1
|
Bhardwaj S, Badiyal A, Dhiman S, Bala J, Walia A. Exploring Halophiles for Reclamation of Saline Soils: Biotechnological Interventions for Sustainable Agriculture. J Basic Microbiol 2025:e70048. [PMID: 40357706 DOI: 10.1002/jobm.70048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/22/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025]
Abstract
Soil salinization is a major constraint on agricultural productivity, particularly in arid and semi-arid regions where limited rainfall cannot wash salts from plant root zones. This leads to disruptions in water uptake, ion balance, photosynthesis, respiration, nutrient absorption, hormone regulation and rhizosphere microbiome disturbances in plants. Chemical and biological methods can help mitigate soil salinity, but biological approaches, like using halophytes and salt-tolerant microorganisms, are preferred for environmental sustainability. Halophytes, however, represent only about 1% of flora and are habitat specific, so halophilic plant growth-promoting (PGP) microbes have emerged as a key eco-friendly solution. Halophilic PGP bacteria have shown promise in remediating saline soils, enhancing fertility and boosting crop resilience by inducing salinity tolerance (IST) and promoting plant growth traits. In the era of modern agriculture where chemical inputs are at their peak of application rendering the soil infertile, halophilic PGP bacteria represent a promising, sustainable approach to support food security, aligning with Sustainable Development Goals for zero hunger.
Collapse
Affiliation(s)
- Shiwani Bhardwaj
- Department of Microbiology, College of Basic Sciences, CSK Himachal Pradesh Agricultural University, Palampur, India
| | - Anila Badiyal
- Department of Microbiology, College of Basic Sciences, CSK Himachal Pradesh Agricultural University, Palampur, India
| | - Shailja Dhiman
- Department of Plant Breeding and Genetics, College of Agriculture, CSK Himachal Pradesh Agricultural University, Palampur, India
| | - Jyoti Bala
- Department of Organic Agriculture and Natural Farming, College of Agriculture, CSK Himachal Pradesh Agricultural University, Palampur, India
| | - Abhishek Walia
- Department of Microbiology, College of Basic Sciences, CSK Himachal Pradesh Agricultural University, Palampur, India
| |
Collapse
|
2
|
Ramesh R, Rekha ND, Gopal S. Pseudomonas aeruginosa biofilm: treatment strategies to combat infection. Arch Microbiol 2025; 207:141. [PMID: 40348909 DOI: 10.1007/s00203-025-04346-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/25/2025] [Accepted: 04/26/2025] [Indexed: 05/14/2025]
Abstract
Pseudomonas aeruginosa is an opportunistic human pathogenic bacterium that is a common cause of both acute and chronic infections. Multidrug-resistant P. aeruginosa poses a significant challenge to antibiotics and therapeutic approaches due to its pathogenicity, virulence, and biofilm-forming ability mediated by quorum sensing. Understanding the pathogenic mechanisms is essential for developing potential drug targets. In this regard, strategies aimed at combating the targeted inhibition of virulence, quorum sensing pathways, secretion systems, biofilm-associated two-component systems, and signalling system regulators (such as c-di-GMP) associated with biofilm formation are critical. Several new antimicrobial agents have been developed using these strategies, including antimicrobial peptides, bacteriophages, nanoantibiotics, photodynamics, and natural products, which are considered promising therapeutic tools. In this review, we address the concept of biofilms, their regulation, and recent treatment strategies to target P. aeruginosa, a clinically significant pathogen known for biofilm formation.
Collapse
Affiliation(s)
- Rashmi Ramesh
- Department of Studies in Microbiology, University of Mysore, Manasagangotri, Mysuru, Karnataka, India
| | - N D Rekha
- Department of Biotechnology, JSS College of Arts, Commerce and Science (Autonomous), Mysuru, Karnataka, India
| | - Shubha Gopal
- Department of Studies in Microbiology, University of Mysore, Manasagangotri, Mysuru, Karnataka, India.
| |
Collapse
|
3
|
Matthews EL, Hirsch MJ, Prokopczuk F, Jones LI, Martínez E, Barnes JW, Krick S. Wound repair and immune function in the Pseudomonas infected CF lung: before and after highly effective modulator therapy. Front Cell Infect Microbiol 2025; 15:1566495. [PMID: 40357395 PMCID: PMC12066499 DOI: 10.3389/fcimb.2025.1566495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/04/2025] [Indexed: 05/15/2025] Open
Abstract
The leading cause of death for people with cystic fibrosis (pwCF) continues to be due to respiratory-related illnesses. Both wound repair and immune cell responses are dysregulated in the CF airways, creating a cycle of unresolved injury and perpetuating inflammation. PwCF are predisposed to colonization and infections with opportunistic bacteria like Pseudomonas aeruginosa (Pa), the most common adult pathogen in CF. Pa possesses key virulence factors that can exacerbate chronic inflammation and lung injury. With the approval of highly effective modulator therapies like elexacaftor/tezacaftor/ivacaftor (ETI), pwCF eligible for ETI have seen drastic improvements in lung function and clinical outcomes, including an increased life expectancy. While modulator therapies are improving bronchial epithelial cellular processes in wound repair and some areas of immunity, many of these processes do not reach a non-CF baseline state or have not been thoroughly studied. The effect of modulator therapy on Pa may lead to a reduction in infection, but in more longitudinal studies, there is not always eradication of Pa, and colonization and infection frequency can return to pre-modulator levels over time. Finally, in this review we explore the current state of additional treatments for CF lung disease, independent of CFTR genotype, including anti-inflammatories, phage-therapies, and Pa vaccines.
Collapse
Affiliation(s)
- Emma Lea Matthews
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Meghan June Hirsch
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Federico Prokopczuk
- Department of Microbiology, The University of Alabama at Birmingham,
Birmingham, AL, United States
| | - Luke I. Jones
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eriel Martínez
- Department of Microbiology, The University of Alabama at Birmingham,
Birmingham, AL, United States
| | - Jarrod W. Barnes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Stefanie Krick
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
4
|
Okpala OE, Rondevaldova J, Kokoska L. Anti-inflammatory drugs as potential antimicrobial agents: a review. Front Pharmacol 2025; 16:1557333. [PMID: 40264668 PMCID: PMC12011823 DOI: 10.3389/fphar.2025.1557333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/13/2025] [Indexed: 04/24/2025] Open
Abstract
The association and causal role of infectious agents in chronic inflammatory diseases have major implications for public health, treatment, and prevention. Pharmacological treatment of combined infectious and inflammatory diseases requires the administration of multiple drugs, including antibiotics and anti-inflammatory drugs. However, this can cause adverse effects, and therefore, dual-action drugs need to be developed. Anti-inflammatory drugs that have already shown antimicrobial properties appear to be promising candidates. NSAIDs, namely aceclofenac, diclofenac, and ibuprofen, were tested in clinical trials with patients diagnosed with uncomplicated urinary tract infections (UTIs) and cellulitis. The administration of ibuprofen, a drug tested in the highest number of studies, resulted in symptom resolution in patients with UTIs. Additionally, ibuprofen caused a high survival rate in mice infected with Pseudomonas aeruginosa and demonstrated potent in vitro antibacterial effects against Bacillus cereus, Escherichia coli, and Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA) (MIC 0.625-2.5 mg/L). For most anti-inflammatory drugs, only data showing their in vitro and in vivo antimicrobial effects are available. Among these, auranofin caused a high survival rate in mice infected with Enterococcus faecium, S. aureus, and Clostridioides difficile. It also produced a strong in vitro growth-inhibitory effect against Streptococcus agalactiae, S. pneumoniae, S. aureus, S. epidermidis, Bacillus subtilis, C. difficile, E. faecalis, E. faecium, and Mycobacterium tuberculosis (MIC 0.0015-5 mg/L). Similarly, aspirin caused a high survival rate in M. tuberculosis-infected mice and strong to moderate in vitro activity against E. coli, B. cereus, P. aeruginosa, Enterobacter aerogenes, Klebsiella pneumoniae and Salmonella choleraesuis (MIC 1.2-5 mg/L). Moreover, topical application of celecoxib resulted in a high reduction in MRSA burden in mice. However, it only caused moderate in vitro effects against S. epidermidis, S. aureus and Bacillus subitilis (MIC 16-64 mg/L). These data suggest that certain non-steroidal anti-inflammatory drugs (NSAIDs) are promising drug candidates for the development of dual-action drugs for the potential treatment of combined infectious and inflammatory diseases such as tuberculosis, musculoskeletal infections and UTIs. Nevertheless, future clinical trials must be conducted to ascertain the antibacterial effect of these NSAIDs before their practical use.
Collapse
Affiliation(s)
| | | | - Ladislav Kokoska
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Prague, Czechia
| |
Collapse
|
5
|
Bie S, Yuan H, Shi C, Li C, Lu M, Yao Z, Liu R, Lu D, Ma T, Yu H. Antibiofilm activity of Plumbagin against Staphylococcus aureus. Sci Rep 2025; 15:7948. [PMID: 40055436 PMCID: PMC11889106 DOI: 10.1038/s41598-025-92435-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/27/2025] [Indexed: 05/13/2025] Open
Abstract
In chronic infections caused by Staphylococcus aureus, biofilm is a major virulence factor. In Staphylococcus aureus biofilms, bacteria are embedded in a matrix of extracellular polymeric substances and are highly tolerant to antimicrobial drugs. However, the lack of effective solutions to inhibit biofilm formation remains a challenge, and the mechanism of inhibition of biofilm formation targeting extracellular polymeric substances is unclear. The aim of the present study was to investigate the inhibitory mechanisms of Plumbagin against Staphylococcus aureus biofilms formation by affecting secretion of extracellular polymeric substances using the high-content screening. Our results showed Plumbagin (16 µg/mL) inhibited biofilm formation, revealing a significant reduction in both biomass and bacterial metabolic activity, and disrupted the biofilm structure, leading to a significant decrease in both biological volume and average thickness (P ≤ 0.01). High-content screening imaging indicated that the Plumbagin treatment induced alterations in the extracellular polymeric substances of Staphylococcus aureus biofilm, significantly reducing the quantities of extracellular polysaccharide, proteins and extracellular DNA. Interestingly, extracellular DNA within the matrix was found to be the most sensitive to Plumbagin treatment. Extracellular DNA formation was significantly inhibited at a concentration of 4 µg/mL, whereas the inhibition of extracellular polysaccharide and proteins required a higher concentration of 8 µg/mL. Overall, these results demonstrated the inhibitory effects of Plumbagin on Staphylococcus aureus biofilm formation and extracellular polymeric substances secretion, suggesting that extracellular DNA may be a potential target for the anti-biofilm activity of Plumbagin. These findings will provide new insights into the mode of action of Plumbagin in treating infections caused by Staphylococcus aureus biofilms.
Collapse
Affiliation(s)
- Songtao Bie
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Intelligent and Green Pharmaceuticals for Traditional Chinese Medicine, Tianjin, China.
| | - Hui Yuan
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Chen Shi
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Chunshuang Li
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Ming Lu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Ze Yao
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Ruobing Liu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Ding Lu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Tenglong Ma
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Heshui Yu
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| |
Collapse
|
6
|
Weaver AA, Shrout JD. Use of analytical strategies to understand spatial chemical variation in bacterial surface communities. J Bacteriol 2025; 207:e0040224. [PMID: 39873490 PMCID: PMC11841061 DOI: 10.1128/jb.00402-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
Not only do surface-growing microbes such as biofilms display specific traits compared to planktonic cells, but also they display many heterogeneous behaviors over many spatial and temporal contexts. While the application of molecular genetics tools to extract or visualize gene expression or regulatory function data is now common in studying surface growth, the use of analytical chemistry tools to visualize the spatiotemporal distribution of chemical products synthesized by these surface microbes is less common. Here, we review chemical imaging tools that have been used to inform our understanding of surface-growing microbes. We highlight the use of confocal Raman Microscopy, surface-enhanced Raman spectroscopy, matrix-assisted laser desorption/ionization, secondary ion mass spectrometry, desorption electrospray ionization, and electrochemical imaging that have been applied to assess two-dimensional chemical profiles of bacteria. We specifically discuss the use of these tools to study rhamnolipids, alkylquinolones, and phenazines of the bacterium Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Abigail A. Weaver
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | - Joshua D. Shrout
- Department of Civil and Environmental Engineering and Earth Sciences, University of Notre Dame, Notre Dame, Indiana, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
7
|
Schandl S, Osondu-Chuka G, Guagliano G, Perak S, Petrini P, Briatico-Vangosa F, Reimhult E, Guillaume O. Acetylation of alginate enables the production of inks that mimic the chemical properties of P. aeruginosa biofilm. J Mater Chem B 2025; 13:2796-2809. [PMID: 39871625 PMCID: PMC11773326 DOI: 10.1039/d4tb02675f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
The reason why certain bacteria, e.g., Pseudomonas aeruginosa (PA), produce acetylated alginate (Alg) in their biofilms remains one of the most intriguing facts in microbiology. Being the main structural component of the secreted biofilm, like the one formed in the lungs of cystic fibrosis (CF) patients, Alg plays a crucial role in protecting the bacteria from environmental stress and potential threats. Nonetheless, to investigate the PA biofilm environment and its lack of susceptibility to antibiotic treatment, the currently developed in vitro biofilm models use native seaweed Alg, which is a non-acetylated Alg. The role of the acetyl side group on the backbone of bacterial Alg has never been elucidated, and the transposition of experimental results obtained from such systems to clinical conditions (e.g., to treat CF-infection) may be hazardous. We systematically investigated the influence of acetylation on the physico-chemical and mechanical properties of Alg in solution and Ca2+-crosslinked hydrogels. Furthermore, we assessed how the acetylation influenced the interaction of Alg with tobramycin, a common aminoglycoside antibiotic for PA. Our study revealed that the degree of acetylation directly impacts the viscosity and Young's Modulus of Alg in a pH-dependent manner. Acetylation increased the mesh size in biofilm-like Alg hydrogels, directly influencing antibiotic penetration. Our results provide essential insights to create more clinically relevant in vitro infection models to test the efficacy of new drugs or to better understand the 3D microenvironment of PA biofilms.
Collapse
Affiliation(s)
- Stephan Schandl
- Institute of Materials Science and Technology, Technische Universität Wien, Vienna, Austria.
- Austrian Cluster of Tissue Regeneration, Austria
| | - Goodness Osondu-Chuka
- Department of Bionanosciences, Institute of Colloid and Biointerface Science, BOKU University, Austria
| | - Giuseppe Guagliano
- Department of Chemistry, Materials and Chemical Engineering 'G Natta', Politecnico di Milano, Milan, Italy
| | - Stjepan Perak
- Institute of Materials Science and Technology, Technische Universität Wien, Vienna, Austria.
| | - Paola Petrini
- Department of Chemistry, Materials and Chemical Engineering 'G Natta', Politecnico di Milano, Milan, Italy
| | - Francesco Briatico-Vangosa
- Department of Chemistry, Materials and Chemical Engineering 'G Natta', Politecnico di Milano, Milan, Italy
| | - Erik Reimhult
- Department of Bionanosciences, Institute of Colloid and Biointerface Science, BOKU University, Austria
| | - Olivier Guillaume
- Institute of Materials Science and Technology, Technische Universität Wien, Vienna, Austria.
- Austrian Cluster of Tissue Regeneration, Austria
| |
Collapse
|
8
|
Akoulina EA, Bonartseva GA, Dudun AA, Kochevalina MY, Bonartsev AP, Voinova VV. Current State of Research on the Mechanisms of Biological Activity of Alginates. BIOCHEMISTRY. BIOKHIMIIA 2025; 90:S263-S286. [PMID: 40164162 DOI: 10.1134/s0006297924604519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/02/2024] [Accepted: 10/31/2024] [Indexed: 04/02/2025]
Abstract
Alginates are anionic unbranched plant and bacterial polysaccharides composed of mannuronic and guluronic acid residues. Alginates can form hydrogels under mild conditions in the presence of divalent cations (e.g., Ca2+). Because of their capacity to form gels, high biocompatibility, and relatively low cost, these polysaccharides are employed in pharmaceutical industry, medicine, food industry, cosmetology, and agriculture. Alginate oligomers produced by enzymatic cleavage of high-molecular-weight algal alginates are used as medicinal agents and dietary supplements. The global market for alginate-based products exceeds $1 billion. Alginates and their oligomers have attracted a special interest in biomedical sciences due to manifestation of various types of therapeutic activity. Across more than 50-year history of studies of alginates, over 60% scientific articles in this field have been published in the last 5 years. Unfortunately, the works dedicated to the mechanisms of biological activity of alginates and their oligosaccharides are still very scarce. This review analyzes the current state of research on the mechanisms (mainly biochemical) underlying biological and therapeutic activities of alginates (antioxidant, antibacterial, anti-inflammatory, antitumor, neuroprotective, antihypertensive, regenerative, and prebiotic). A comprehensive understanding of these mechanisms will not only improve the efficiency of alginate application in medicine and other traditional fields (cosmetology, food industry), but might also reveal their potential in new areas such as tissue engineering, nanobiotechnology, and bioelectronics.
Collapse
Affiliation(s)
- Elizaveta A Akoulina
- Biological Faculty, Shenzhen MSU-BIT University, Shenzhen, Guangdong Province, 518172, China
| | - Garina A Bonartseva
- Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Science, Moscow, 119071, Russia
| | - Andrey A Dudun
- Federal Research Centre "Fundamentals of Biotechnology", Russian Academy of Science, Moscow, 119071, Russia
- Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, 123098, Russia
| | | | - Anton P Bonartsev
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - Vera V Voinova
- Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, 123098, Russia.
| |
Collapse
|
9
|
Khalaf MM, Gouda M, Abou Taleb MF, Heakal FET, Abd El-Lateef HM. Fabrication of smart nanogel based on carrageenan and green coffee extract as a long-term antifouling agent to improve biofilm prevention in food production. Food Chem 2024; 461:140719. [PMID: 39146677 DOI: 10.1016/j.foodchem.2024.140719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/22/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024]
Abstract
This study investigates the extract of the bioactive compounds from green coffee extract (GCE) and the loading of two different concentrations of GCE (1% and 2%) onto carrageenan nanogels (CAR NGs) to compare their antibacterial and antibiofilm effects with unloaded nanogels (NGs). The bioactive compounds of GCE were characterized using GC-MS analysis. The GCE1 and GCE2 were successfully deposited onto the surface of CAR NGs. The antibacterial and antibiofilm potential of prepared NGs were conducted against some foodborne pathogens (E. coli O157, Salmonella enterica, Staphylococcus aureus, and Listeria monocytogenes). The results of GC-MS analysis indicated that there were identified 16 bioactive compounds in GCE, including caffeine (36.27%), Dodemorph (9.04%), and D-Glycero-d-ido-heptose (2.44%), contributing to its antimicrobial properties. The antibacterial coatings demonstrated a notable antimicrobial effect, showing zone of inhibition (ZOI) diameters of up to 37 mm for GCE2 loaded CAR NGs. The minimum inhibitory concentration (MIC) values for GCE2 loaded CAR NGs were 80 ppm for E. coli O157, and 120 ppm for S. enterica, S. aureus, and L. monocytogenes, achieving complete bacterial inactivation within 10-15 min of exposure. Both GCE1 and GCE2 loaded CAR NGs significantly reduced biofilm cell densities on stainless steel (SS) materials for E. coli O157, S. enterica, S. aureus, and L. monocytogenes, with reductions ranging from 60% to 95%. Specifically, biofilm densities were reduced by up to 95% for E. coli O157, 89% for S. enterica, 85% for S. aureus, and 80% for L. monocytogenes. Results of the toxicity evaluation indicated that the NGs were non-toxic and biocompatible, with predicted EC50 values proved their biocompatibility and safety. These results recommended that GCE loaded CAR NGs are promising as natural antimicrobial agents for enhancing food safety and extending shelf life. Further, the study concluded that incorporating GCE into CAR NGs is an effective strategy for developing sustainable antimicrobial coatings for the food industry and manufacturing.
Collapse
Affiliation(s)
- Mai M Khalaf
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia; Department of Chemistry, Faculty of Science, Sohag University, Sohag 82524, Egypt.
| | - Mohamed Gouda
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia.
| | - Manal F Abou Taleb
- Department of Chemistry, College of Science and Humanities, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | | | - Hany M Abd El-Lateef
- Department of Chemistry, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia; Department of Chemistry, Faculty of Science, Sohag University, Sohag 82524, Egypt
| |
Collapse
|
10
|
Yao S, Yang H, Zhang M, Xian J, Zhou R, Jin Y, Huang J, Wu C. Sucrose contributed to the biofilm formation of Tetragenococcus halophilus and changed the biofilm structure. Food Microbiol 2024; 124:104616. [PMID: 39244368 DOI: 10.1016/j.fm.2024.104616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 09/09/2024]
Abstract
Based on the previous research results that the addition of sucrose in the medium improved the biofilm formation of Tetragenococcus halophilus, the influence of sucrose on biofilm formation was explored. Moreover, the influence of exogenous expression of related genes sacA and galE from T. halophilus on the biofilm formation of L. lactis NZ9000 was investigated. The results showed that the addition of sucrose in the medium improved the biofilm formation, the resistance of biofilm cells to freeze-drying stress, and the contents of exopolysaccharides (EPS) and eDNA in the T. halophilus biofilms. Meanwhile, the addition of sucrose in the medium changed the monosaccharide composition of EPS and increased the proportion of glucose and galactose in the monosaccharide composition. Under 2.5% (m/v) salt stress condition, the expression of gene sacA promoted the biofilm formation and the EPS production of L. lactis NZ9000 with the sucrose addition in the medium and changed the EPS monosaccharide composition. The expression of gene galE up-regulated the proportion of rhamnose, galactose, and arabinose in the monosaccharide composition of EPS, and down-regulated the proportion of glucose and mannose. This study will provide a theoretical basis for regulating the biofilm formation of T. halophilus, and provide a reference for the subsequent research on lactic acid bacteria biofilms.
Collapse
Affiliation(s)
- Shangjie Yao
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China; GuiZhou XiJiu Co., Ltd, Xishui, Guizhou, 564622, China
| | - Huan Yang
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Min Zhang
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Jiao Xian
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China; GuiZhou XiJiu Co., Ltd, Xishui, Guizhou, 564622, China
| | - Rongqing Zhou
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Yao Jin
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Jun Huang
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Chongde Wu
- College of Biomass Science and Engineering, Sichuan University, Chengdu, 610065, China.
| |
Collapse
|
11
|
Ragupathi H, Pushparaj MM, Gopi SM, Govindarajan DK, Kandaswamy K. Biofilm matrix: a multifaceted layer of biomolecules and a defensive barrier against antimicrobials. Arch Microbiol 2024; 206:432. [PMID: 39402397 DOI: 10.1007/s00203-024-04157-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/24/2024] [Accepted: 10/03/2024] [Indexed: 11/10/2024]
Abstract
Bacterial cells often exist in the form of sessile aggregates known as biofilms, which are polymicrobial in nature and can produce slimy Extracellular Polymeric Substances (EPS). EPS is often referred to as a biofilm matrix and is a heterogeneous mixture of various biomolecules such as polysaccharides, proteins, and extracellular DNA/RNA (eDNA/RNA). In addition, bacteriophage (phage) was also found to be an integral component of the matrix and can serve as a protective barrier. In recent years, the roles of proteins, polysaccharides, and phages in the virulence of biofilms have been well studied. However, a mechanistic understanding of the release of such biomolecules and their interactions with antimicrobials requires a thorough review. Therefore, this article critically reviews the various mechanisms of release of matrix polymers. In addition, this article also provides a contemporary understanding of interactions between various biomolecules to protect biofilms against antimicrobials. In summary, this article will provide a thorough understanding of the functions of various biofilm matrix molecules.
Collapse
Affiliation(s)
- Harini Ragupathi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Mahamahima Muthuswamy Pushparaj
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Sarves Mani Gopi
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India
| | - Deenadayalan Karaiyagowder Govindarajan
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore, Singapore
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Drive, 637371, Singapore, Singapore
| | - Kumaravel Kandaswamy
- Research Center for Excellence in Microscopy, Department of Biotechnology, Kumaraguru College of Technology, Coimbatore, Tamil Nadu, 641049, India.
| |
Collapse
|
12
|
Zhang P, Zeng P, Lai CKC, Ip M, To KKW, Zuo Z, Xia J, Leung SSY. Synergism of colistin and globular endolysins against multidrug-resistant gram-negative bacteria. Int J Biol Macromol 2024; 278:134670. [PMID: 39151868 DOI: 10.1016/j.ijbiomac.2024.134670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Endolysins (lysins), a novel class of antibacterial agents derived from bacteriophages, efficiently lyse bacteria by degrading the peptidoglycan layer within the bacterial wall. Colistin, a classic peptide antibiotic with the ability to permeabilize the outer membrane, has recently shown great promise in synergizing with lysins against gram-negative bacteria. However, the exact mechanisms responsible for their synergy remain unclear. Here, we first demonstrated the synergistic bacterial killing of various lysin and colistin combinations. With a model lysin, LysAB2, we then confirmed that there is a threshold concentration of colistin causing sufficient permeabilization of the outer membrane for lysin to access the peptidoglycan layer and subsequently exert its lytic ability. The threshold colistin concentrations were found to range 0.2-0.8 μM for the tested bacteria, with the exact value largely depending on the density of lipopolysaccharides on the outer membrane. Beyond the threshold colistin level, LysAB2 could synergize with colistin at a concentration as low as 0.31 μM. Next, we proved for the first time that lysin-induced degradation of the peptidoglycan layer facilitated the disruption of cytoplasmic membrane by colistin, elevated the level of reactive oxygen species in bacterial cells, and boosted the killing effect of colistin. Additionally, the colistin-lysin combination could effectively eliminate established biofilms due to the biofilm dispersal ability of lysin. The in-vivo efficacy was preliminary confirmed in a Galleria mellonella infection model for combination with colistin doses (≥ 1.8 μg/larvae), which could reach beyond the threshold concentration, and a fixed LysAB2 dose (10 μg/larvae). In summary, our study provided the first experimental evidence unravelling the mechanisms behind the synergy of colistin and lysins. All these findings provided important insights in guiding the dosing strategy for applying this combination in future development.
Collapse
Affiliation(s)
- Pengfei Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Ping Zeng
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Christopher K C Lai
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Margaret Ip
- Department of Microbiology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Zhong Zuo
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong.
| | - Sharon S Y Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
13
|
Abril AG, Calo-Mata P, Böhme K, Villa TG, Barros-Velázquez J, Sánchez-Pérez Á, Pazos M, Carrera M. Shotgun proteomic analyses of Pseudomonas species isolated from fish products. Food Chem 2024; 450:139342. [PMID: 38631198 DOI: 10.1016/j.foodchem.2024.139342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/25/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
Numerous Pseudomonas species can infect aquatic animals, such as farmed rainbow trout, sea trout, sea bass, and sea bream, by causing disease or stress reactions. In aquaculture facilities, a number of Pseudomonas species have been isolated and identified as the main pathogens. The present study describes the characterization of 18 Pseudomonas strains, isolated from fish products using shotgun proteomics. The bacterial proteomes obtained were further analyzed to identify the main functional pathway proteins involved. In addition, this study revealed the presence of 1015 non-redundant peptides related to virulence factors. An additional 25 species-specific peptides were identified as putative Pseudomonas spp. biomarkers. The results constitute the largest dataset, described thus far for the rapid identification and characterization of Pseudomonas species present in edible fish; furthermore, these data can provide the basis for further research into the development of new therapies against these harmful pathogens.
Collapse
Affiliation(s)
- Ana G Abril
- Department of Food Technology, Marine Research Institute (IIM-CSIC), Spanish National Research Council (CSIC), 36208 Vigo, Spain; Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Santiago de Compostela, 15898 Santiago de Compostela, Spain.
| | - Pilar Calo-Mata
- Department of Analytical Chemistry, Nutrition and Food Science, Food Technology Division, School of Veterinary Sciences, University of Santiago de Compostela, Campus Lugo, 27002 Lugo, Spain.
| | - Karola Böhme
- Department of Analytical Chemistry, Nutrition and Food Science, Food Technology Division, School of Veterinary Sciences, University of Santiago de Compostela, Campus Lugo, 27002 Lugo, Spain
| | - Tomás G Villa
- Department of Microbiology and Parasitology, Faculty of Pharmacy, University of Santiago de Compostela, 15898 Santiago de Compostela, Spain
| | - Jorge Barros-Velázquez
- Department of Analytical Chemistry, Nutrition and Food Science, Food Technology Division, School of Veterinary Sciences, University of Santiago de Compostela, Campus Lugo, 27002 Lugo, Spain.
| | - Ángeles Sánchez-Pérez
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, NSW 2006, Australia
| | - Manuel Pazos
- Department of Food Technology, Marine Research Institute (IIM-CSIC), Spanish National Research Council (CSIC), 36208 Vigo, Spain.
| | - Mónica Carrera
- Department of Food Technology, Marine Research Institute (IIM-CSIC), Spanish National Research Council (CSIC), 36208 Vigo, Spain.
| |
Collapse
|
14
|
Motta EVS, de Jong TK, Gage A, Edwards JA, Moran NA. Glyphosate effects on growth and biofilm formation in bee gut symbionts and diverse associated bacteria. Appl Environ Microbiol 2024; 90:e0051524. [PMID: 39012136 PMCID: PMC11337805 DOI: 10.1128/aem.00515-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/22/2024] [Indexed: 07/17/2024] Open
Abstract
Biofilm formation is a common adaptation enabling bacteria to thrive in various environments and withstand external pressures. In the context of host-microbe interactions, biofilms play vital roles in establishing microbiomes associated with animals and plants and are used by opportunistic microbes to facilitate survival within hosts. Investigating biofilm dynamics, composition, and responses to environmental stressors is crucial for understanding microbial community assembly and biofilm regulation in health and disease. In this study, we explore in vivo colonization and in vitro biofilm formation abilities of core members of the honey bee (Apis mellifera) gut microbiota. Additionally, we assess the impact of glyphosate, a widely used herbicide with antimicrobial properties, and a glyphosate-based herbicide formulation on growth and biofilm formation in bee gut symbionts as well as in other biofilm-forming bacteria associated with diverse animals and plants. Our results demonstrate that several strains of core bee gut bacterial species can colonize the bee gut, which probably depends on their ability to form biofilms. Furthermore, glyphosate exposure elicits variable effects on bacterial growth and biofilm formation. In some instances, the effects correlate with the bacteria's ability to encode a susceptible or tolerant version of the enzyme inhibited by glyphosate in the shikimate pathway. However, in other instances, no such correlation is observed. Testing the herbicide formulation further complicates comparisons, as results often diverge from glyphosate exposure alone, suggesting that co-formulants influence bacterial growth and biofilm formation. These findings highlight the nuanced impacts of environmental stressors on microbial biofilms, with both ecological and host health-related implications. IMPORTANCE Biofilms are essential for microbial communities to establish and thrive in diverse environments. In the honey bee gut, the core microbiota member Snodgrassella alvi forms biofilms, potentially aiding the establishment of other members and promoting interactions with the host. In this study, we show that specific strains of other core members, including Bifidobacterium, Bombilactobacillus, Gilliamella, and Lactobacillus, also form biofilms in vitro. We then examine the impact of glyphosate, a widely used herbicide that can disrupt the bee microbiota, on bacterial growth and biofilm formation. Our findings demonstrate the diverse effects of glyphosate on biofilm formation, ranging from inhibition to enhancement, reflecting observations in other beneficial or pathogenic bacteria associated with animals and plants. Thus, glyphosate exposure may influence bacterial growth and biofilm formation, potentially shaping microbial establishment on host surfaces and impacting health outcomes.
Collapse
Affiliation(s)
- Erick V. S. Motta
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
- Department of Entomology, Texas A&M University, College Station, Texas, USA
| | - Tyler K. de Jong
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Alejandra Gage
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Joseph A. Edwards
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, Texas, USA
| | - Nancy A. Moran
- Department of Integrative Biology, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
15
|
Ding L, Wang G, Wang J, Peng Y, Cai S, Khan SU, Cui Z, Zhang X, Wu C, Smyth H. Targeted treatment for biofilm-based infections using PEGylated tobramycin. J Control Release 2024; 372:43-58. [PMID: 38866243 DOI: 10.1016/j.jconrel.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/24/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
Chronic infections often involve biofilm-based bacteria, in which the biofilm results in significant resistance against antimicrobial agents and prevents eradication of the infection. The physicochemical barrier presented by the biofilm matrix is a major impediment to the delivery of many antibiotics. Previously, PEGylation has been shown to improve antibiotic penetration into biofilms in vitro. In these studies, PEGylating tobramycin was investigated both in vitro and in vivo. Two distinct PEGylated tobramycin molecules were synthesized (mPEG-SA-Tob and mPEG-AA-Tob). Then, in a P. aeruginosa biofilm in vitro model, we found that mPEG-SA-Tob can operate as a prodrug and showed 7 times more effectiveness than tobramycin (MIC80: 14 μM vs.100 μM). This improved biofilm eradication is attributable to the fact that mPEG-SA-Tob can aid tobramycin to penetrate through the biofilm and overcome the alginate-mediated antibiotic resistance. Finally, we used an in vivo biofilm-based chronic pulmonary infection rat model to confirm the therapeutic impact of mPEG-SA-Tob on biofilm-based chronic lung infection. mPEG-SA-Tob has a better therapeutic impact than tobramycin in that it cannot only stop P. aeruginosa from multiplying in the lungs but can also reduce inflammation caused by infections and prevent a recurrence infection. Overall, our findings show that PEGylated tobramycin is an effective treatment for biofilm-based chronic lung infections.
Collapse
Affiliation(s)
- Li Ding
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Guanlin Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, Guangdong, PR China
| | - Jieliang Wang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ying Peng
- Division of Chemical Biology and Medicinal Chemistry, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Shihao Cai
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Shafi Ullah Khan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia; INSERM U1086 ANTICIPE (Interdisciplinary Research Unit for Cancers Prevention and Treatment), BioTICLA Laboratory (Precision Medicine for Ovarian Cancers), Université de Caen Normandie, Caen, France; Comprehensive Cancer Center François Baclesse, UNICANCER, Caen, France
| | - Zhengrong Cui
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA
| | - Xuejuan Zhang
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 511443, China.
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou, Guangdong 511443, China
| | - Hugh Smyth
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
16
|
D’Aquila P, De Rose E, Sena G, Scorza A, Cretella B, Passarino G, Bellizzi D. Quorum Quenching Approaches against Bacterial-Biofilm-Induced Antibiotic Resistance. Antibiotics (Basel) 2024; 13:619. [PMID: 39061301 PMCID: PMC11273524 DOI: 10.3390/antibiotics13070619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
With the widespread phenomenon of antibiotic resistance and the diffusion of multiple drug-resistant bacterial strains, enormous efforts are being conducted to identify suitable alternative agents against pathogenic microorganisms. Since an association between biofilm formation and antibiotic resistance phenotype has been observed, a promising strategy pursued in recent years focuses on controlling and preventing this formation by targeting and inhibiting the Quorum Sensing (QS) system, whose central role in biofilm has been extensively demonstrated. Therefore, the research and development of Quorum Quenching (QQ) compounds, which inhibit QS, has gradually attracted the attention of researchers and has become a new strategy for controlling harmful microorganisms. Among these, a number of both natural and synthetic compounds have been progressively identified as able to interrupt the intercellular communication within a microbial community and the adhesion to a surface, thus disintegrating mature/preformed biofilms. This review describes the role played by QS in the formation of bacterial biofilms and then focuses on the mechanisms of different natural and synthetic QS inhibitors (QSIs) exhibiting promising antibiofilm ability against Gram-positive and Gram-negative bacterial pathogens and on their applications as biocontrol strategies in various fields.
Collapse
Affiliation(s)
- Patrizia D’Aquila
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Elisabetta De Rose
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Giada Sena
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Angelo Scorza
- Villa Ermelinda, Progetto Terza Età, 88842 Cutro, Italy; (A.S.); (B.C.)
| | | | - Giuseppe Passarino
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| | - Dina Bellizzi
- Department of Biology, Ecology and Earth Sciences, University of Calabria, 87036 Rende, Italy; (P.D.); (E.D.R.); (G.S.); (G.P.)
| |
Collapse
|
17
|
Prosswimmer T, Nick SE, Bryers JD, Daggett V. Designed De Novo α-Sheet Peptides Destabilize Bacterial Biofilms and Increase the Susceptibility of E. coli and S. aureus to Antibiotics. Int J Mol Sci 2024; 25:7024. [PMID: 39000131 PMCID: PMC11241457 DOI: 10.3390/ijms25137024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/16/2024] Open
Abstract
Biofilm-associated microbes are 10-1000 times less susceptible to antibiotics. An emerging treatment strategy is to target the structural components of biofilm to weaken the extracellular matrix without introducing selective pressure. Biofilm-associated bacteria, including Escherichia coli and Staphylococcus aureus, generate amyloid fibrils to reinforce their extracellular matrix. Previously, de novo synthetic α-sheet peptides designed in silico were shown to inhibit amyloid formation in multiple bacterial species, leading to the destabilization of their biofilms. Here, we investigated the impact of inhibiting amyloid formation on antibiotic susceptibility. We hypothesized that combined administration of antibiotics and α-sheet peptides would destabilize biofilm formation and increase antibiotic susceptibility. Two α-sheet peptides, AP90 and AP401, with the same sequence but inverse chirality at every amino acid were tested: AP90 is L-amino acid dominant while AP401 is D-amino acid dominant. For E. coli, both peptides increased antibiotic susceptibility and decreased the biofilm colony forming units when administered with five different antibiotics, and AP401 caused a greater increase in all cases. For S. aureus, increased biofilm antibiotic susceptibility was also observed for both peptides, but AP90 outperformed AP401. A comparison of the peptide effects demonstrates how chirality influences biofilm targeting of gram-negative E. coli and gram-positive S. aureus. The observed increase in antibiotic susceptibility highlights the role amyloid fibrils play in the reduced susceptibility of bacterial biofilms to specific antibiotics. Thus, the co-administration of α-sheet peptides and existing antibiotics represents a promising strategy for the treatment of biofilm infections.
Collapse
Affiliation(s)
- Tatum Prosswimmer
- Molecular Engineering Program, University of Washington, Seattle, WA 98195, USA
| | - Sarah E. Nick
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| | - James D. Bryers
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| | - Valerie Daggett
- Molecular Engineering Program, University of Washington, Seattle, WA 98195, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA;
| |
Collapse
|
18
|
Niepa THR, Locke LW, Corcoran TE, Lee JS. Editorial: Mechanobiology of biofilms and associated host -pathogen interactions. Front Cell Infect Microbiol 2024; 14:1416131. [PMID: 38716197 PMCID: PMC11074442 DOI: 10.3389/fcimb.2024.1416131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 06/19/2024] Open
Affiliation(s)
- Tagbo H R Niepa
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Landon W Locke
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, United States
| | - Timothy E Corcoran
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Janet S Lee
- Division of Pulmonary and Critical Care Medicine, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
19
|
Behzadnia A, Moosavi-Nasab M, Oliyaei N. Anti-biofilm activity of marine algae-derived bioactive compounds. Front Microbiol 2024; 15:1270174. [PMID: 38680918 PMCID: PMC11055458 DOI: 10.3389/fmicb.2024.1270174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
A large number of microbial species tend to communicate and produce biofilm which causes numerous microbial infections, antibiotic resistance, and economic problems across different industries. Therefore, advanced anti-biofilms are required with novel attributes and targets, such as quorum sensing communication system. Meanwhile, quorum sensing inhibitors as promising anti-biofilm molecules result in the inhibition of particular phenotype expression blocking of cell-to-cell communication, which would be more acceptable than conventional strategies. Many natural products are identified as anti-biofilm agents from different plants, microorganisms, and marine extracts. Marine algae are promising sources of broadly novel compounds with anti-biofilm activity. Algae extracts and their metabolites such as sulfated polysaccharides (fucoidan), carotenoids (zeaxanthin and lutein), lipid and fatty acids (γ-linolenic acid and linoleic acid), and phlorotannins can inhibit the cell attachment, reduce the cell growth, interfere in quorum sensing pathway by blocking related enzymes, and disrupt extracellular polymeric substances. In this review, the mechanisms of biofilm formation, quorum sensing pathway, and recently identified marine algae natural products as anti-biofilm agents will be discussed.
Collapse
Affiliation(s)
- Asma Behzadnia
- Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Marzieh Moosavi-Nasab
- Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| | - Najmeh Oliyaei
- Seafood Processing Research Center, School of Agriculture, Shiraz University, Shiraz, Iran
- Department of Food Science and Technology, School of Agriculture, Shiraz University, Shiraz, Iran
| |
Collapse
|
20
|
Zhang L, Bera H, Guo Y, Shi C, Ulrik Lind J, Radeke C, Wang J, Wang H, Zhao X, Cun D, Yang M. Co-spray dried inhalable composite powders of ciprofloxacin and alginate oligosaccharide as anti-biofilm therapy. Int J Pharm 2024; 654:123949. [PMID: 38417723 DOI: 10.1016/j.ijpharm.2024.123949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/25/2024] [Accepted: 02/25/2024] [Indexed: 03/01/2024]
Abstract
The treatment of chronic respiratory infections caused by biofilm formation are extremely challenging owing to poor drug penetration into the complex biofilm structure and high drug resistance. Local delivery of an antibiotic together with a non-antibiotic adjuvant to the lungs could often enhance the therapeutic responses by targeting different bacterial growth pathways and minimizing drug resistance. In this study, we designed new inhalable dry powders containing ciprofloxacin (CIP) and OligoG (Oli, a low-molecular-weight alginate oligosaccharide impairing the mucoid biofilms by interacting with their cationic ions) to combat respiratory bacterial biofilm infections. The resulting powders were characterized with respect to their morphology, solid-state property, surface chemistry, moisture sorption behavior, and dissolution rate. The aerosol performance and storage stability of the dry powders were also evaluated. The results showed that inhalable dry powders composed of CIP and Oli could be readily accomplished via the wet milling and spray drying process. Upon the storage under 20 ± 2 °C/20 ± 2 % relative humidity (RH) for one month, there was no significant change in the in vitro aerosol performances of the dry powders. In contrast, the dry powders became non-inhalable following the storage at 20 ± 2 °C/53 ± 2 % RH for one month due to the hygroscopic nature of Oli, which could be largely prevented by incorporation of leucine. Collectively, this study suggests that the newly developed co-spray-dried powders composed of CIP and Oli might represent a promising and alternative treatment strategy against respiratory bacterial biofilm infections.
Collapse
Affiliation(s)
- Li Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, No. 103, 110016 Shenyang, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; Lanzhou Institute of Biological Products Co., Ltd, Lanzhou, China
| | - Hriday Bera
- Dr. B.C. Roy College of Pharmacy & Allied Health Sciences, Durgapur, India
| | - Yi Guo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, No. 103, 110016 Shenyang, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; Taiyuan Health School, Taiyuan, China
| | - Changzhi Shi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, No. 103, 110016 Shenyang, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Johan Ulrik Lind
- Department of Health Technology (DTU Health Tech), Technical University of Denmark, Lyngby, Denmark
| | - Carmen Radeke
- Department of Health Technology (DTU Health Tech), Technical University of Denmark, Lyngby, Denmark
| | - Junwei Wang
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark
| | - Hengzhuang Wang
- Department of Clinical Microbiology, Copenhagen University Hospital, Rigshospitalet, Henrik Harpestrengsvej 4A, DK-2100 Copenhagen, Denmark; Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Xia Zhao
- Key Laboratory of Marine Drugs, Ministry of Education, Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Dongmei Cun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, No. 103, 110016 Shenyang, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China.
| | - Mingshi Yang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Wenhua Road, No. 103, 110016 Shenyang, China; Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China; Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen, Denmark.
| |
Collapse
|
21
|
Yang Y, Kong X, Niu B, Yang J, Chen Q. Differences in Biofilm Formation of Listeria monocytogenes and Their Effects on Virulence and Drug Resistance of Different Strains. Foods 2024; 13:1076. [PMID: 38611380 PMCID: PMC11011679 DOI: 10.3390/foods13071076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Listeria monocytogenes is recognized as one of the primary pathogens responsible for foodborne illnesses. The ability of L. monocytogenes to form biofilms notably increases its resistance to antibiotics such as ampicillin and tetracycline, making it exceedingly difficult to eradicate. Residual bacteria within the processing environment can contaminate food products, thereby posing a significant risk to public health. In this study, we used crystal violet staining to assess the biofilm-forming capacity of seven L. monocytogenes strains and identified ATCC 19112 as the strain with the most potent biofilm-forming. Subsequent fluorescence microscopy observations revealed that the biofilm-forming capacity was markedly enhanced after two days of culture. Then, we investigated into the factors contributing to biofilm formation and demonstrated that strains with more robust extracellular polymer secretion and self-agglutination capabilities exhibited a more pronounced ability to form biofilms. No significant correlation was found between surface hydrophobicity and biofilm formation capability. In addition, we found that after biofilm formation, the adhesion and invasion of cells were enhanced and drug resistance increased. Therefore, we hypothesized that the formation of biofilm makes L. monocytogenes more virulent and more difficult to remove by antibiotics. Lastly, utilizing RT-PCR, we detected the expression levels of genes associated with biofilm formation, including those involved in quorum sensing (QS), flagellar synthesis, and extracellular polymer production. These genes were significantly upregulated after biofilm formation. These findings underscore the critical relationship between extracellular polymers, self-agglutination abilities, and biofilm formation. In conclusion, the establishment of biofilms not only enhances L. monocytogenes' capacity for cell invasion and adhesion but also significantly increases its resistance to drugs, presenting a substantial threat to food safety.
Collapse
Affiliation(s)
- Yujuan Yang
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.Y.); (B.N.)
| | - Xiangxiang Kong
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China;
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Bing Niu
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.Y.); (B.N.)
| | - Jielin Yang
- Technical Centre for Animal, Plant and Food Inspection and Quarantine of Shanghai Customs, Shanghai 200135, China
| | - Qin Chen
- School of Life Sciences, Shanghai University, Shanghai 200444, China; (Y.Y.); (B.N.)
| |
Collapse
|
22
|
Iaconis A, De Plano LM, Caccamo A, Franco D, Conoci S. Anti-Biofilm Strategies: A Focused Review on Innovative Approaches. Microorganisms 2024; 12:639. [PMID: 38674584 PMCID: PMC11052202 DOI: 10.3390/microorganisms12040639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Biofilm (BF) can give rise to systemic infections, prolonged hospitalization times, and, in the worst case, death. This review aims to provide an overview of recent strategies for the prevention and destruction of pathogenic BFs. First, the main phases of the life cycle of BF and maturation will be described to identify potential targets for anti-BF approaches. Then, an approach acting on bacterial adhesion, quorum sensing (QS), and the extracellular polymeric substance (EPS) matrix will be introduced and discussed. Finally, bacteriophage-mediated strategies will be presented as innovative approaches against BF inhibition/destruction.
Collapse
Affiliation(s)
- Antonella Iaconis
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Domenico Franco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Sabrina Conoci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy
- URT Lab Sens Beyond Nano—CNR-DSFTM, Department of Physical Sciences and Technologies of Matter, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
23
|
Sousa AM, Ferreira D, Rodrigues LR, Pereira MO. Aptamer-based therapy for fighting biofilm-associated infections. J Control Release 2024; 367:522-539. [PMID: 38295992 DOI: 10.1016/j.jconrel.2024.01.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/06/2024] [Accepted: 01/27/2024] [Indexed: 02/06/2024]
Abstract
Biofilms are key players in the pathogenesis of most of chronic infections associated with host tissue or fluids and indwelling medical devices. These chronic infections are hard to be treated due to the increased biofilms tolerance towards antibiotics in comparison to planktonic (or free living) cells. Despite the advanced understanding of their formation and physiology, biofilms continue to be a challenge and there is no standardized therapeutic approach in clinical practice to eradicate them. Aptamers offer distinctive properties, including excellent affinity, selectivity, stability, making them valuable tools for therapeutic purposes. This review explores the flexibility and designability of aptamers as antibiofilm drugs but, importantly, as targeting tools for diverse drug and delivery systems. It highlights specific examples of application of aptamers in biofilms of diverse species according to different modes of action including inhibition of motility and adhesion, blocking of quorum sensing molecules, and dispersal of biofilm-cells to planktonic state. Moreover, it discusses the limitations and challenges that impaired an increased success of the use of aptamers on biofilm management, as well as the opportunities related to aptamers modifications that can significantly expand their applicability on the biofilm field.
Collapse
Affiliation(s)
- Ana Margarida Sousa
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| | - Débora Ferreira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Lígia Raquel Rodrigues
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB - Centre of Biological Engineering, University of Minho, 4710-057 Braga, Portugal; LABBELS - Associate Laboratory, Braga, Guimarães, Portugal.
| |
Collapse
|
24
|
Rouillard KR, Esther CP, Kissner WJ, Plott LM, Bowman DW, Markovetz MR, Hill DB. Combination treatment to improve mucociliary transport of Pseudomonas aeruginosa biofilms. PLoS One 2024; 19:e0294120. [PMID: 38394229 PMCID: PMC10890754 DOI: 10.1371/journal.pone.0294120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/25/2023] [Indexed: 02/25/2024] Open
Abstract
People with muco-obstructive pulmonary diseases such as cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) often have acute or chronic respiratory infections that are difficult to treat due in part to the accumulation of hyperconcentrated mucus within the airway. Mucus accumulation and obstruction promote chronic inflammation and infection and reduce therapeutic efficacy. Bacterial aggregates in the form of biofilms exhibit increased resistance to mechanical stressors from the immune response (e.g., phagocytosis) and chemical treatments including antibiotics. Herein, combination treatments designed to disrupt the mechanical properties of biofilms and potentiate antibiotic efficacy are investigated against mucus-grown Pseudomonas aeruginosa biofilms and optimized to 1) alter biofilm viscoelastic properties, 2) increase mucociliary transport rates, and 3) reduce bacterial viability. A disulfide bond reducing agent (tris(2-carboxyethyl)phosphine, TCEP), a surfactant (NP40), a biopolymer (hyaluronic acid, HA), a DNA degradation enzyme (DNase), and an antibiotic (tobramycin) are tested in various combinations to maximize biofilm disruption. The viscoelastic properties of biofilms are quantified with particle tracking microrheology and transport rates are quantified in a mucociliary transport device comprised of fully differentiated primary human bronchial epithelial cells. The combination of the NP40 with hyaluronic acid and tobramycin was the most effective at increasing mucociliary transport rates, decreasing the viscoelastic properties of mucus, and reducing bacterial viability. Multimechanistic targeting of biofilm infections may ultimately result in improved clinical outcomes, and the results of this study may be translated into future in vivo infection models.
Collapse
Affiliation(s)
- Kaitlyn R. Rouillard
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | | | - William J. Kissner
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | - Lucas M. Plott
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | - Dean W. Bowman
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | - Matthew R. Markovetz
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
| | - David B. Hill
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC, United States of America
- Joint Department of Biomedical Engineering, UNC Chapel Hill, Chapel Hill, NC, United States of America
| |
Collapse
|
25
|
Böhning J, Tarafder AK, Bharat TA. The role of filamentous matrix molecules in shaping the architecture and emergent properties of bacterial biofilms. Biochem J 2024; 481:245-263. [PMID: 38358118 PMCID: PMC10903470 DOI: 10.1042/bcj20210301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/16/2024]
Abstract
Numerous bacteria naturally occur within spatially organised, multicellular communities called biofilms. Moreover, most bacterial infections proceed with biofilm formation, posing major challenges to human health. Within biofilms, bacterial cells are embedded in a primarily self-produced extracellular matrix, which is a defining feature of all biofilms. The biofilm matrix is a complex, viscous mixture primarily composed of polymeric substances such as polysaccharides, filamentous protein fibres, and extracellular DNA. The structured arrangement of the matrix bestows bacteria with beneficial emergent properties that are not displayed by planktonic cells, conferring protection against physical and chemical stresses, including antibiotic treatment. However, a lack of multi-scale information at the molecular level has prevented a better understanding of this matrix and its properties. Here, we review recent progress on the molecular characterisation of filamentous biofilm matrix components and their three-dimensional spatial organisation within biofilms.
Collapse
Affiliation(s)
- Jan Böhning
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| | - Abul K. Tarafder
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| | - Tanmay A.M. Bharat
- Structural Studies Division, MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, U.K
| |
Collapse
|
26
|
Scoffone VC, Barbieri G, Irudal S, Trespidi G, Buroni S. New Antimicrobial Strategies to Treat Multi-Drug Resistant Infections Caused by Gram-Negatives in Cystic Fibrosis. Antibiotics (Basel) 2024; 13:71. [PMID: 38247630 PMCID: PMC10812592 DOI: 10.3390/antibiotics13010071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/23/2024] Open
Abstract
People with cystic fibrosis (CF) suffer from recurrent bacterial infections which induce inflammation, lung tissue damage and failure of the respiratory system. Prolonged exposure to combinatorial antibiotic therapies triggers the appearance of multi-drug resistant (MDR) bacteria. The development of alternative antimicrobial strategies may provide a way to mitigate antimicrobial resistance. Here we discuss different alternative approaches to the use of classic antibiotics: anti-virulence and anti-biofilm compounds which exert a low selective pressure; phage therapies that represent an alternative strategy with a high therapeutic potential; new methods helping antibiotics activity such as adjuvants; and antimicrobial peptides and nanoparticle formulations. Their mechanisms and in vitro and in vivo efficacy are described, in order to figure out a complete landscape of new alternative approaches to fight MDR Gram-negative CF pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Silvia Buroni
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, 27100 Pavia, Italy; (V.C.S.); (G.B.); (S.I.); (G.T.)
| |
Collapse
|
27
|
Dehari D, Kumar DN, Chaudhuri A, Kumar A, Kumar R, Kumar D, Singh S, Nath G, Agrawal AK. Bacteriophage entrapped chitosan microgel for the treatment of biofilm-mediated polybacterial infection in burn wounds. Int J Biol Macromol 2023; 253:127247. [PMID: 37802451 DOI: 10.1016/j.ijbiomac.2023.127247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/19/2023] [Accepted: 10/02/2023] [Indexed: 10/10/2023]
Abstract
Staphylococcus aureus (S. aureus) and Pseudomonas aeruginosa (P. aeruginosa) bacteria are most commonly present in burn wound infections. Multidrug resistance (MDR) and biofilm formation make it difficult to treat these infections. Bacteriophages (BPs) are proven as an effective therapy against MDR as well as biofilm-associated wound infections. In the present work, a naturally inspired bacteriophage cocktail loaded chitosan microparticles-laden topical gel has been developed for the effective treatment of these infections. Bacteriophages against MDR S. aureus (BPSAФ1) and P. aeruginosa (BPPAФ1) were isolated and loaded separately and in combination into the chitosan microparticles (BPSAФ1-CHMPs, BPPAФ1-CHMPs, and MBP-CHMPs), which were later incorporated into the SEPINEO™ P 600 gel (BPSAФ1-CHMPs-gel, BPPAФ1-CHMPs-gel, and MBP-CHMPs-gel). BPs were characterized for their morphology, lytic activity, burst size, and hemocompatibility, and BPs belongs to Caudoviricetes class. Furthermore, BPSAФ1-CHMPs, BPPAФ1-CHMPs, and MBP-CHMPs had an average particle size of 1.19 ± 0.11, 1.42 ± 0.21, and 2.84 ± 0.28 μm, respectively, and expressed promising in vitro antibiofilm eradication potency. The ultrasound and photoacoustic imaging in infected burn wounds demonstrated improved wound healing reduced inflammation and increased oxygen saturation following treatment with BPs formulations. The obtained results suggested that the incorporation of the BPs in the MP-gel protected the BPs, sustained the BPs release, and improved the antibacterial activity.
Collapse
Affiliation(s)
- Deepa Dehari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Dulla Naveen Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Aiswarya Chaudhuri
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Akshay Kumar
- Department of Microbiology, Institute of Medial Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Rajesh Kumar
- Department of Microbiology, Institute of Medial Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India
| | - Sanjay Singh
- Babasaheb Bhimrao Ambedkar University, Lucknow 226025, U.P., India
| | - Gopal Nath
- Department of Microbiology, Institute of Medial Science, Banaras Hindu University, Varanasi 221005, U.P., India
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, U.P., India.
| |
Collapse
|
28
|
Song Y, Sun M, Mu G, Tuo Y. Exopolysaccharide produced by Lactiplantibacillus plantarum Y12 exhibits inhibitory effect on the Shigella flexneri genes expression related to biofilm formation. Int J Biol Macromol 2023; 253:127048. [PMID: 37748596 DOI: 10.1016/j.ijbiomac.2023.127048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/27/2023]
Abstract
Shigella is a specific enteric pathogen in humans, causing symptoms of bacterial dysentery. The biofilm formation of S. flexneri contributes to the emergence of multidrug resistance and facilitates the establishment of persistent chronic infections. This study investigated the regulatory effects of Lactiplantibacillus plantarum Y12 exopolysaccharide (L-EPS) on gene expression and its spatial hindrance effects in inhibiting the biofilm formation of S. flexneri. The transcriptome analysis revealed a significant impact of L-EPS on the gene expression profile of S. flexneri, with a total of 968 genes showing significant changes (507 up-regulated and 461 down-regulated). The significantly down-regulated KEGG metabolic pathway enriched in phosphotransferase system, Embden-Meyerhf-Parnas, Citrate cycle, Lipopolysaccharide biosynthesis, Cationic antimicrobial peptide resistance, Two-component system. Moreover, L-EPS significantly down-regulated the gene expression levels of fimbriae synthesis (fimF), lipopolysaccharide synthesis (lptE, lptB), anchor protein repeat domain (arpA), virulence factor (lpp, yqgB), antibiotic resistance (marR, cusB, mdtL, mdlB), heavy metal resistance (zraP), and polysaccharide synthesis (mtgA, mdoB, mdoC). The expression of biofilm regulator factor (bssS) and two-component system suppressor factor (mgrB) were significantly up-regulated. The RT-qPCR results indicated that a major component of L-EPS (L-EPS 2-1) exhibited the gene regulatory effect on the S. flexneri biofilm formation. Furthermore, electrophoresis and isothermal microtitration calorimetry demonstrated that the interaction between L-EPS 2-1 and eDNA is electrostatic dependent on the change in environmental pH, disrupting the stable spatial structure of S. flexneri biofilm. In conclusion, L-EPS inhibited the biofilm formation of S. flexneri through gene regulation and spatial obstruction effects.
Collapse
Affiliation(s)
- Yinglong Song
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China
| | - Mengying Sun
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China
| | - Guangqing Mu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China; Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian 116034, PR China.
| | - Yanfeng Tuo
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
29
|
Guo Q, Zhan Y, Zhang W, Wang J, Yan Y, Wang W, Lin M. Development and Regulation of the Extreme Biofilm Formation of Deinococcus radiodurans R1 under Extreme Environmental Conditions. Int J Mol Sci 2023; 25:421. [PMID: 38203592 PMCID: PMC10778927 DOI: 10.3390/ijms25010421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
To grow in various harsh environments, extremophiles have developed extraordinary strategies such as biofilm formation, which is an extremely complex and progressive process. However, the genetic elements and exact mechanisms underlying extreme biofilm formation remain enigmatic. Here, we characterized the biofilm-forming ability of Deinococcus radiodurans in vitro under extreme environmental conditions and found that extremely high concentrations of NaCl or sorbitol could induce biofilm formation. Meantime, the survival ability of biofilm cells was superior to that of planktonic cells in different extreme conditions, such as hydrogen peroxide stress, sorbitol stress, and high UV radiation. Transcriptome profiles of D. radiodurans in four different biofilm development stages further revealed that only 13 matched genes, which are involved in environmental information processing, carbohydrate metabolism, or stress responses, share sequence homology with genes related to the biofilm formation of Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus. Overall, 64% of the differentially expressed genes are functionally unknown, indicating the specificity of the regulatory network of D. radiodurans. The mutation of the drRRA gene encoding a response regulator strongly impaired biofilm formation ability, implying that DrRRA is an essential component of the biofilm formation of D. radiodurans. Furthermore, transcripts from both the wild type and the drRRA mutant were compared, showing that the expression of drBON1 (Deinococcus radioduransBON domain-containing protein 1) significantly decreased in the drRRA mutant during biofilm development. Further analysis revealed that the drBON1 mutant lacked the ability to form biofilm and DrRRA, and as a facilitator of biofilm formation, could directly stimulate the transcription of the biofilm-related gene drBON1. Overall, our work highlights a molecular mechanism mediated by the response regulator DrRRA for controlling extreme biofilm formation and thus provides guidance for future studies to investigate novel mechanisms that are used by D. radiodurans to adapt to extreme environments.
Collapse
Affiliation(s)
- Qiannan Guo
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yuhua Zhan
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wei Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jin Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yongliang Yan
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wenxiu Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Min Lin
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (Q.G.); (Y.Z.); (W.Z.); (J.W.); (Y.Y.); (W.W.)
- Key Laboratory of Agricultural Microbiome (MARA), Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
30
|
Saad MH, Sidkey NM, El-Fakharany EM. Identification and statistical optimization of a novel alginate polymer extracted from newly isolated Synechocystis algini MNE ON864447 with antibacterial activity. Microb Cell Fact 2023; 22:229. [PMID: 37932753 PMCID: PMC10629183 DOI: 10.1186/s12934-023-02240-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/28/2023] [Indexed: 11/08/2023] Open
Abstract
Cyanobacteria are a potential source of promising secondary metabolites with different biological activities, including antibacterial, antiviral, antifungal, antiprotozoal, and anticancer activities. To combat the emergence of antibiotic resistance, there is an urgent requirement for new drugs, and cyanobacteria metabolites can constitute alternative new antibacterial medication. The chemical complexity of their exopolysaccharides indicates that they have the potential to be bioactive molecules with many biological activities. The present study aimed to produce and optimise a novel alginate polymer from a newly isolated cyanobacterium, S. algini MNE ON864447, in addition to its promising antibacterial activity. We successfully isolated a new cyanobacterium strain, S. algini MNE ON864447 from the Nile River, which produces alginate as an extracellular polymeric substance. The isolated cyanobacterial alginate was identified using a set of tests, including FTIR, TLC, HPLC, GC-MS, and 1H NMR. Plackett-Burman statistical design showed that working volume (X1), the incubation period (X2), and inoculum size (X3) are the most significant variables affecting the production of alginate. The highest alginate production (3.57 g/L) was obtained using 4% inoculum size in 400 mL medium/L conical flask after 20 days of the incubation period. The extracted alginate showed potent antibacterial activity against both Gram-negative and Gram-positive bacteria and Streptococcus mutants (NCTC10449) are the most sensitive tested pathogen for purified cyanobacterial alginate with inhibition zone diameters of 34 ± 0.1 mm at 10 mg/mL of purified alginate while Vibro cholera (NCTC 8021) the lowest sensitive one and showed inhibition zone diameters of 22.5 ± 0.05 mm at the same cyanobacterial alginate concentration. This antibacterial activity is a critical step in the development of antibacterial drugs and presents a new challenge to fight against multi-resistant bacteria.
Collapse
Affiliation(s)
- Mabroka H Saad
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research andTechnological Applications (SRTA-City), New Borg AL Arab, Alexandria, Egypt
- Botany & Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Cairo, Egypt
| | - Nagwa M Sidkey
- Botany & Microbiology Department, Faculty of Science, Al-Azhar University (Girls Branch), Cairo, Egypt
| | - Esmail M El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research andTechnological Applications (SRTA-City), New Borg AL Arab, Alexandria, Egypt.
- Pharmaceutical and Fermentation Industries Development Centre (PFIDC), City of Scientific Research and Technological Applications (SRTA-City), New Borg Al-Arab, Alexandria, Egypt.
| |
Collapse
|
31
|
Guo Y, Mao Z, Ran F, Sun J, Zhang J, Chai G, Wang J. Nanotechnology-Based Drug Delivery Systems to Control Bacterial-Biofilm-Associated Lung Infections. Pharmaceutics 2023; 15:2582. [PMID: 38004561 PMCID: PMC10674810 DOI: 10.3390/pharmaceutics15112582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/09/2023] [Accepted: 10/17/2023] [Indexed: 11/26/2023] Open
Abstract
Airway mucus dysfunction and impaired immunological defenses are hallmarks of several lung diseases, including asthma, cystic fibrosis, and chronic obstructive pulmonary diseases, and are mostly causative factors in bacterial-biofilm-associated respiratory tract infections. Bacteria residing within the biofilm architecture pose a complex challenge in clinical settings due to their increased tolerance to currently available antibiotics and host immune responses, resulting in chronic infections with high recalcitrance and high rates of morbidity and mortality. To address these unmet clinical needs, potential anti-biofilm therapeutic strategies are being developed to effectively control bacterial biofilm. This review focuses on recent advances in the development and application of nanoparticulate drug delivery systems for the treatment of biofilm-associated respiratory tract infections, especially addressing the respiratory barriers of concern for biofilm accessibility and the various types of nanoparticles used to combat biofilms. Understanding the obstacles facing pulmonary drug delivery to bacterial biofilms and nanoparticle-based approaches to combatting biofilm may encourage researchers to explore promising treatment modalities for bacterial-biofilm-associated chronic lung infections.
Collapse
Affiliation(s)
- Yutong Guo
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Zeyuan Mao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fang Ran
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Jingfeng Zhang
- The Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo 315000, China
| | - Guihong Chai
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jian Wang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| |
Collapse
|
32
|
Zhai H, Yeo J. Controlling biofilm transport with porous metamaterials designed with Bayesian learning. J Mech Behav Biomed Mater 2023; 147:106127. [PMID: 37797554 DOI: 10.1016/j.jmbbm.2023.106127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/29/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
Biofilm growth and transport in confined systems frequently occur in natural and engineered systems. Designing customizable engineered porous materials for controllable biofilm transportation properties could significantly improve the rapid utilization of biofilms as engineered living materials for applications in pollution alleviation, material self-healing, energy production, and many more. We combine Bayesian optimization (BO) and individual-based modeling to conduct design optimizations for maximizing different porous materials' (PM) biofilm transportation capability. We first characterize the acquisition function in BO for designing 2-dimensional porous membranes. We use the expected improvement acquisition function for designing lattice metamaterials (LM) and 3-dimensional porous media (3DPM). We find that BO is 92.89% more efficient than the uniform grid search method for LM and 223.04% more efficient for 3DPM. For all three types of structures, the selected characterization simulation tests are in good agreement with the design spaces approximated with Gaussian process regression. All the extracted optimal designs exhibit better biofilm growth and transportability than unconfined space without substrates. Our comparison study shows that PM stimulates biofilm growth by taking up volumetric space and pushing biofilms' upward growth, as evidenced by a 20% increase in bacteria cell numbers in unconfined space compared to porous materials, and 128% more bacteria cells in the target growth region for PM-induced biofilm growth compared with unconfined growth. Our work provides deeper insights into the design of substrates to tune biofilm growth, analyzing the optimization process and characterizing the design space, and understanding biophysical mechanisms governing the growth of biofilms.
Collapse
Affiliation(s)
- Hanfeng Zhai
- Sibley School of Mechanical and Aerospace Engineering Cornell University, Ithaca, NY 14850, USA
| | - Jingjie Yeo
- Sibley School of Mechanical and Aerospace Engineering Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
33
|
Zhang A, Cao Z, Zhao L, Zhang Q, Fu L, Li J, Liu T. Characterization of bifunctional alginate lyase Aly644 and antimicrobial activity of enzymatic hydrolysates. Appl Microbiol Biotechnol 2023; 107:6845-6857. [PMID: 37698609 DOI: 10.1007/s00253-023-12745-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/02/2023] [Accepted: 08/11/2023] [Indexed: 09/13/2023]
Abstract
An alginate lyase gene aly644 encoding a member of polysaccharide lyase family 6 was obtained from a metagenome of Antarctic macroalgae-associated microbes. The gene was expressed heterologously in Escherichia coli, and the recombinant protein was purified using a Ni-NTA His Tag Kit. With sodium alginate as the substrate, recombinant Aly644 exhibited an optimum reaction temperature of 50°C and an optimum reaction pH of 7.0. The Vmax and Km values of Aly644 toward sodium alginate were 112.36 mg/mL·min and 16.75 mg/mL, respectively. Substrate specificity analysis showed that Aly644 was a bifunctional alginate lyase that hydrolyzed both polyguluronic acid and polymannuronic acid. The hydrolysis products of Aly644 with sodium alginate as the substrate were detected by thin-layer chromatography, and were mainly di- and trisaccharides. The oligosaccharides produced by degradation of sodium alginate by Aly644 inhibited the mycelial growth of the plant pathogens Phytophthora capsici and Fulvia fulva; the 50% maximal effective concentration (EC50) values were 297.45 and 452.89 mg/L, and the 90% maximal effective concentration (EC90) values were 1341.45 and 2693.83 mg/L, respectively. This highlights that Aly644 is a potential candidate enzyme for the industrial production of alginate oligosaccharides with low degree of polymerization. Enzyme-hydrolyzed alginate oligosaccharides could support the development of green agriculture as natural antimicrobial agents. KEY POINTS: • An alginate lyase was obtained from a metagenome of Antarctic macroalgae-associated microbes. • Aly644 is a bifunctional alginate lyase with excellent thermostability and pH stability. • The enzymatic hydrolysates of Aly644 directly inhibited Phytophthora capsici and Fulvia fulva.
Collapse
Affiliation(s)
- Ao Zhang
- Department of Pharmaceutical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China
| | - Zhe Cao
- Key Lab of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| | - Luying Zhao
- Key Lab of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| | - Qian Zhang
- Key Lab of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| | - Liping Fu
- Key Lab of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China
| | - Jiang Li
- Key Lab of Ecological Environment Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China.
| | - Tao Liu
- Department of Pharmaceutical Engineering, College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China.
| |
Collapse
|
34
|
Lv X, Jiang J, Ren J, Li H, Yang D, Song X, Hu Y, Wang W, Dong X. Nitric Oxide-Assisted Photodynamic Therapy for Enhanced Penetration and Hypoxic Bacterial Biofilm Elimination. Adv Healthc Mater 2023; 12:e2302031. [PMID: 37515529 DOI: 10.1002/adhm.202302031] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/24/2023] [Indexed: 07/31/2023]
Abstract
The presence of a biofilm matrix barrier and hypoxic microenvironment within the biofilm significantly impedes the efficacy of photodynamic therapy for bacterial biofilm infections. Herein, a phototherapeutic nanoagent with type-I photodynamic behavior and nitric oxide (NO) release performance is reported for overcoming biofilm-associated infectious diseases. Sodium nitroprusside (SNP), a NO donor, is loaded onto amino-modified mesoporous silica nanoparticles (MSN) to form MSN@SNP NPs. The resulting nanoparticles are further modified with a porphyrin-based metal-organic framework (Ti-TCPP MOF) to obtain MSN@MOF/SNP NPs (MMS NPs) for phototherapeutic applications. In the hypoxia biofilm microenvironment, the MMS NPs release NO to enhance the biofilm permeability and induce the generation of hydroxyl radical (•OH) and superoxide anion radical (O2 •- ) via Type-I photodynamic pathway under laser irradiation. Subsequently, the biofilm-associated infections are effectively eliminated through reactive oxygen species (ROS) and NO gas synergistic therapy. In addition, NO also stimulates collagen deposition and promotes angiogenesis in vivo. Therefore, the MMS NPs efficiently treat biofilm-related infections, providing an alternative approach to combat biofilm-associated infectious diseases.
Collapse
Affiliation(s)
- Xinyi Lv
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Jingai Jiang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Jie Ren
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Hui Li
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Dongliang Yang
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Xuejiao Song
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Yanling Hu
- College of life and health, Nanjing Polytechnic Institute, Nanjing, 210048, China
| | - Wenjun Wang
- School of Physical Science and Information Technology, Liaocheng University, Liaocheng, 252059, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), School of Physical and Mathematical Sciences, Nanjing Tech University (NanjingTech), Nanjing, 211816, China
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, China
| |
Collapse
|
35
|
Bonincontro G, Scuderi SA, Marino A, Simonetti G. Synergistic Effect of Plant Compounds in Combination with Conventional Antimicrobials against Biofilm of Staphylococcus aureus, Pseudomonas aeruginosa, and Candida spp. Pharmaceuticals (Basel) 2023; 16:1531. [PMID: 38004397 PMCID: PMC10675371 DOI: 10.3390/ph16111531] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Bacterial and fungal biofilm has increased antibiotic resistance and plays an essential role in many persistent diseases. Biofilm-associated chronic infections are difficult to treat and reduce the efficacy of medical devices. This global problem has prompted extensive research to find alternative strategies to fight microbial chronic infections. Plant bioactive metabolites with antibiofilm activity are known to be potential resources to alleviate this problem. The phytochemical screening of some medicinal plants showed different active groups, such as stilbenes, tannins, alkaloids, terpenes, polyphenolics, flavonoids, lignans, quinones, and coumarins. Synergistic effects can be observed in the interaction between plant compounds and conventional drugs. This review analyses and summarises the current knowledge on the synergistic effects of plant metabolites in combination with conventional antimicrobials against biofilms of Staphylococcus aureus, Pseudomonas aeruginosa, and Candida albicans. The synergism of conventional antimicrobials with plant compounds can modify and inhibit the mechanisms of acquired resistance, reduce undesirable effects, and obtain an appropriate therapeutic effect at lower doses. A deeper knowledge of these combinations and of their possible antibiofilm targets is needed to develop next-generation novel antimicrobials and/or improve current antimicrobials to fight drug-resistant infections attributed to biofilm.
Collapse
Affiliation(s)
- Graziana Bonincontro
- Department of Environmental Biology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Roma, Italy;
| | - Sarah Adriana Scuderi
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98100 Messina, Italy;
| | - Andreana Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D’Alcontres, 98100 Messina, Italy;
| | - Giovanna Simonetti
- Department of Environmental Biology, Sapienza University of Rome, P.le Aldo Moro, 5, 00185 Roma, Italy;
| |
Collapse
|
36
|
Shu Z, Wang G, Liu F, Xu Y, Sun J, Hu Y, Dong H, Zhang J. Genome Sequencing-Based Mining and Characterization of a Novel Alginate Lyase from Vibrio alginolyticus S10 for Specific Production of Disaccharides. Mar Drugs 2023; 21:564. [PMID: 37999388 PMCID: PMC10672080 DOI: 10.3390/md21110564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/25/2023] Open
Abstract
Alginate oligosaccharides prepared by alginate lyases attracted great attention because of their desirable biological activities. However, the hydrolysis products are always a mixture of oligosaccharides with different degrees of polymerization, which increases the production cost because of the following purification procedures. In this study, an alginate lyase, Alg4755, with high product specificity was identified, heterologously expressed, and characterized from Vibrio alginolyticus S10, which was isolated from the intestine of sea cucumber. Alg4755 belonged to the PL7 family with two catalytic domains, which was composed of 583 amino acids. Enzymatic characterization results show that the optimal reaction temperature and pH of Alg4755 were 35 °C and 8.0, respectively. Furthermore, Alg4755 was identified to have high thermal and pH stability. Moreover, the final hydrolysis products of sodium alginate catalyzed by Alg4755 were mainly alginate disaccharides with a small amount of alginate trisaccharides. The results demonstrate that alginate lyase Alg4755 could have a broad application prospect because of its high product specificity and desirable catalytic properties.
Collapse
Affiliation(s)
- Zhiqiang Shu
- Department of Food Science and Technology, Shanghai Ocean University, Shanghai 200120, China;
- Shandong Marine Resource and Environment Research Institute, Yantai 264006, China; (G.W.)
| | - Gongming Wang
- Shandong Marine Resource and Environment Research Institute, Yantai 264006, China; (G.W.)
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Yantai 264006, China
| | - Fang Liu
- Shandong Marine Resource and Environment Research Institute, Yantai 264006, China; (G.W.)
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Yantai 264006, China
| | - Yingjiang Xu
- Shandong Marine Resource and Environment Research Institute, Yantai 264006, China; (G.W.)
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Yantai 264006, China
| | - Jianan Sun
- Qingdao Key Laboratory of Food Biotechnology, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China; (J.S.)
- Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, China
| | - Yang Hu
- Qingdao Key Laboratory of Food Biotechnology, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China; (J.S.)
- Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, China
| | - Hao Dong
- Qingdao Key Laboratory of Food Biotechnology, College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China; (J.S.)
- Key Laboratory of Biological Processing of Aquatic Products, China National Light Industry, Qingdao 266404, China
| | - Jian Zhang
- Shandong Marine Resource and Environment Research Institute, Yantai 264006, China; (G.W.)
- Yantai Key Laboratory of Quality and Safety Control and Deep Processing of Marine Food, Yantai 264006, China
| |
Collapse
|
37
|
Esfahani MB, Khodavandi A, Alizadeh F, Bahador N. Antibacterial and Anti-Biofilm Activities of Microbial Synthesized Silver and Magnetic Iron Oxide Nanoparticles Against Pseudomonas aeruginosa. IEEE Trans Nanobioscience 2023; 22:956-966. [PMID: 37071524 DOI: 10.1109/tnb.2023.3268138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
Pseudomonas aeruginosa is a human bacterial pathogen causing devastating diseases and equipped with various virulence factors like biofilm formation. Common antibiotic treatment has limited efficacy for the P. aeruginosa present in biofilms because of the increased resistance. In this study, we focused our attention on the antibacterial and anti-biofilm activities of various microbial synthesized silver (nano-Ag) and magnetic iron oxide (nano-Fe3O4) nanoparticles against clinical isolates of P. aeruginosa that displayed ceftazidime resistance. The nano-Ag and nano-Fe3O4 represented great antibacterial properties. Nano-Ag and nano-Fe3O4 exhibited a reduction in the biofilm formation by P. aeruginosa reference strain as determined by crystal violet and XTT assays and light microscopy method. Among all, nano-Ag-2 and 7 owing to inherent attributes and mechanisms of resistance in the bacterial biofilm, exhibited anti-biofilm efficacy against ceftazidime resistance clinical isolate of P. aeruginosa. Moreover, nano-Ag and nano-Fe3O4 changed the relative expression of biofilm-associated genes, PELA and PSLA in a concentration dependent manner by P. aeruginosa reference strain. As revealed by qRT-PCR, the expression levels of biofilm-associated genes were downregulated in P. aeruginosa biofilms treated with nano-Ag, while selected biofilm-associated genes were low expressed under treated with nano-Fe3O4. Results of the study demonstrate the potential of microbial synthesized nano-Ag-2 and 7 to act as anti-biofilm agents against ceftazidime resistance clinical isolate of P. aeruginosa. Molecular targeting of biofilm-associated genes by nano-Ag and nano-Fe3O4 may be candidate for new therapeutics against P. aeruginosa diseases.
Collapse
|
38
|
Pritchard MF, Powell LC, Adams JYM, Menzies G, Khan S, Tøndervik A, Sletta H, Aarstad O, Skjåk-Bræk G, McKenna S, Buurma NJ, Farnell DJJ, Rye PD, Hill KE, Thomas DW. Structure-Activity Relationships of Low Molecular Weight Alginate Oligosaccharide Therapy against Pseudomonas aeruginosa. Biomolecules 2023; 13:1366. [PMID: 37759766 PMCID: PMC10527064 DOI: 10.3390/biom13091366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/26/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023] Open
Abstract
Low molecular weight alginate oligosaccharides have been shown to exhibit anti-microbial activity against a range of multi-drug resistant bacteria, including Pseudomonas aeruginosa. Previous studies suggested that the disruption of calcium (Ca2+)-DNA binding within bacterial biofilms and dysregulation of quorum sensing (QS) were key factors in these observed effects. To further investigate the contribution of Ca2+ binding, G-block (OligoG) and M-block alginate oligosaccharides (OligoM) with comparable average size DPn 19 but contrasting Ca2+ binding properties were prepared. Fourier-transform infrared spectroscopy demonstrated prolonged binding of alginate oligosaccharides to the pseudomonal cell membrane even after hydrodynamic shear treatment. Molecular dynamics simulations and isothermal titration calorimetry revealed that OligoG exhibited stronger interactions with bacterial LPS than OligoM, although this difference was not mirrored by differential reductions in bacterial growth. While confocal laser scanning microscopy showed that both agents demonstrated similar dose-dependent reductions in biofilm formation, OligoG exhibited a stronger QS inhibitory effect and increased potentiation of the antibiotic azithromycin in minimum inhibitory concentration and biofilm assays. This study demonstrates that the anti-microbial effects of alginate oligosaccharides are not purely influenced by Ca2+-dependent processes but also by electrostatic interactions that are common to both G-block and M-block structures.
Collapse
Affiliation(s)
- Manon F. Pritchard
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Lydia C. Powell
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
- Microbiology and Infectious Disease Group, Swansea University Medical School, Swansea SA2 8PP, UK
| | - Jennifer Y. M. Adams
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Georgina Menzies
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK;
| | - Saira Khan
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Anne Tøndervik
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway; (A.T.); (H.S.)
| | - Håvard Sletta
- Department of Bioprocess Technology, SINTEF Materials and Chemistry, N-7465 Trondheim, Norway; (A.T.); (H.S.)
| | - Olav Aarstad
- Department of Biotechnology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway; (O.A.); (G.S.-B.)
| | - Gudmund Skjåk-Bræk
- Department of Biotechnology, Norwegian University of Science and Technology, N-7491 Trondheim, Norway; (O.A.); (G.S.-B.)
| | - Stephen McKenna
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Niklaas J. Buurma
- Physical Organic Chemistry Centre, School of Chemistry, Cardiff University, Cardiff CF10 3AT, UK;
| | - Damian J. J. Farnell
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - Philip D. Rye
- AlgiPharma AS, Industriveien 33, N-1337 Sandvika, Norway;
| | - Katja E. Hill
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| | - David W. Thomas
- Advanced Therapies Group, School of Dentistry, Cardiff University, Cardiff CF14 4XY, UK; (L.C.P.); (J.Y.M.A.); (S.K.); (S.M.); (D.J.J.F.); (K.E.H.); (D.W.T.)
| |
Collapse
|
39
|
Ruhal R, Ghosh M, Kumar V, Jain D. Mutation of putative glycosyl transferases PslC and PslI confers susceptibility to antibiotics and leads to drastic reduction in biofilm formation in Pseudomonas aeruginosa. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001392. [PMID: 37702709 PMCID: PMC10569066 DOI: 10.1099/mic.0.001392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 08/31/2023] [Indexed: 09/14/2023]
Abstract
Pseudomonas aeruginosa is an opportunistic, multidrug-resistant pathogen capable of adapting to numerous environmental conditions and causing fatal infections in immunocompromised patients. The predominant lifestyle of P. aeruginosa is in the form of biofilms, which are structured communities of bacteria encapsulated in a matrix containing exopolysaccharides, extracellular DNA (eDNA) and proteins. The matrix is impervious to antibiotics, rendering the bacteria tolerant to antimicrobials. P. aeruginosa also produces a plethora of virulence factors such as pyocyanin, rhamnolipids and lipopolysaccharides among others. In this study we present the molecular characterization of pslC and pslI genes, of the exopolysaccharide operon, that code for putative glycosyltransferases. PslC is a 303 amino acid containing putative GT2 glycosyltrasferase, whereas PslI is a 367 aa long protein, possibly functioning as a GT4 glycosyltransferase. Mutation in either of these two genes results in a significant reduction in biofilm biomass with concomitant decline in c-di-GMP levels in the bacterial cells. Moreover, mutation in pslC and pslI dramatically increased susceptibility of P. aeruginosa to tobramycin, colistin and ciprofloxacin. Additionally, these mutations also resulted in an increase in rhamnolipids and pyocyanin formation. We demonstrate that elevated rhamnolipids promote a swarming phenotype in the mutant strains. Together these results highlight the importance of PslC and PslI in the biogenesis of biofilms and their potential as targets for increased antibiotic susceptibility and biofilm inhibition.
Collapse
Affiliation(s)
- Rohit Ruhal
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, India
| | - Moumita Ghosh
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, India
| | - Vineet Kumar
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, India
| | - Deepti Jain
- Transcription Regulation Lab, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, 121001, India
| |
Collapse
|
40
|
Rouillard KR, Esther CP, Kissner WJ, Plott LM, Bowman DW, Markovetz MR, Hill DB. Combination Treatment to Improve Mucociliary Transport of Pseudomonas aeruginosa Biofilms. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553173. [PMID: 37645913 PMCID: PMC10461968 DOI: 10.1101/2023.08.14.553173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
People with muco-obstructive pulmonary diseases such as cystic fibrosis (CF) and chronic obstructive pulmonary disease (COPD) often have acute or chronic respiratory infections that are difficult to treat due in part to the accumulation of hyperconcentrated mucus within the airway. Mucus accumulation and obstruction promote chronic inflammation and infection and reduce therapeutic efficacy. Bacterial aggregates in the form of biofilms exhibit increased resistance to mechanical stressors from the immune response (e.g., phagocytosis) and chemical treatments including antibiotics. Herein, combination treatments designed to disrupt the mechanical properties of biofilms and potentiate antibiotic efficacy are investigated against mucus-grown Pseudomonas aeruginosa biofilms and optimized to 1) alter biofilm viscoelastic properties, 2) increase mucociliary transport rates, and 3) reduce bacterial viability. A disulfide bond reducing agent (tris(2-carboxyethyl)phosphine, TCEP), a surfactant (NP40), a biopolymer (hyaluronic acid, HA), a DNA degradation enzyme (DNase), and an antibiotic (tobramycin) are tested in various combinations to maximize biofilm disruption. The viscoelastic properties of biofilms are quantified with particle tracking microrheology and transport rates are quantified in a mucociliary transport device comprised of fully differentiated primary human bronchial epithelial cells. The combination of the NP40 with hyaluronic acid and tobramycin was the most effective at increasing mucociliary transport rates, decreasing the viscoelastic properties of mucus, and reducing bacterial viability. Multimechanistic targeting of biofilm infections may ultimately result in improved clinical outcomes, and the results of this study may be translated into future in vivo infection models.
Collapse
Affiliation(s)
| | | | | | - Lucas M Plott
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC 27599
| | - Dean W Bowman
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC 27599
| | | | - David B Hill
- Marsico Lung Institute, UNC Chapel Hill, Chapel Hill, NC 27599
- Joint Department of Biomedical Engineering, UNC Chapel Hill, NC 27599
| |
Collapse
|
41
|
Xue W, Pritchard MF, Khan S, Powell LC, Stokniene J, Wu J, Claydon N, Reddell P, Thomas DW, Hill KE. Defining in vitro topical antimicrobial and antibiofilm activity of epoxy-tigliane structures against oral pathogens. J Oral Microbiol 2023; 15:2241326. [PMID: 37534218 PMCID: PMC10392292 DOI: 10.1080/20002297.2023.2241326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/04/2023] [Accepted: 07/20/2023] [Indexed: 08/04/2023] Open
Abstract
Background Peri-implantitis has become an inexorable clinical challenge in implantology. Topical immunomodulatory epoxy-tiglianes (EBCs), derived from the Queensland blushwood tree, which induce remodeling and resolve dermal infection via induction of the inflammasome and biofilm disruption, may offer a novel therapeutic approach. Design In vitro antimicrobial activity of EBC structures (EBC-46, EBC-1013 and EBC-147) against Streptococcus mutans, Aggregatibacter actinomycetemcomitans and Porphyromonas gingivalis in minimum inhibitory concentration, growth curve and permeabilization assays were determined. Antibiofilm activity was assessed using minimum biofilm eradication concentration (MBEC) experiments. Biofilm formation and disruption assays were analyzed using confocal laser scanning microscopy, scanning electron microscopy and direct plate counting. Results The observed antimicrobial efficacy of the tested compounds (EBC-1013 > EBC-46 > EBC-147) was directly related to significant membrane permeabilization and growth inhibition (p < 0.05) against planktonic S. mutans and P. gingivalis. Antibiofilm activity was evident in MBEC assays, with S. mutans biofilm formation assays revealing significantly lower biomass volume and increased DEAD:LIVE cell ratio observed for EBC-1013 (p < 0.05). Furthermore, biofilm disruption assays on titanium discs induced significant biofilm disruption in S. mutans and P. gingivalis (p < 0.05). Conclusions EBC-1013 is a safe, semi-synthetic, compound, demonstrating clear antimicrobial biofilm disruption potential in peri-implantitis.
Collapse
Affiliation(s)
- Wenya Xue
- Advanced Therapies Group, Cardiff School of Dentistry, Heath Park, Cardiff University, Cardiff, UK
| | - Manon F. Pritchard
- Advanced Therapies Group, Cardiff School of Dentistry, Heath Park, Cardiff University, Cardiff, UK
| | - Saira Khan
- Advanced Therapies Group, Cardiff School of Dentistry, Heath Park, Cardiff University, Cardiff, UK
| | - Lydia C. Powell
- Microbiology and Infectious Disease Group, Swansea University Medical School, Swansea, UK
| | - Joana Stokniene
- Advanced Therapies Group, Cardiff School of Dentistry, Heath Park, Cardiff University, Cardiff, UK
| | - Jingxiang Wu
- Advanced Therapies Group, Cardiff School of Dentistry, Heath Park, Cardiff University, Cardiff, UK
| | - Nicholas Claydon
- Advanced Therapies Group, Cardiff School of Dentistry, Heath Park, Cardiff University, Cardiff, UK
| | - Paul Reddell
- QBiotics Group Limited, Yungaburra, Queensland, Australia
| | - David W. Thomas
- Advanced Therapies Group, Cardiff School of Dentistry, Heath Park, Cardiff University, Cardiff, UK
| | - Katja E. Hill
- Advanced Therapies Group, Cardiff School of Dentistry, Heath Park, Cardiff University, Cardiff, UK
| |
Collapse
|
42
|
Coppens B, Belpaire TE, Pešek J, Steenackers HP, Ramon H, Smeets B. Anomalous diffusion of nanoparticles in the spatially heterogeneous biofilm environment. iScience 2023; 26:106861. [PMID: 37260744 PMCID: PMC10227381 DOI: 10.1016/j.isci.2023.106861] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/20/2023] [Accepted: 05/08/2023] [Indexed: 06/02/2023] Open
Abstract
Biofilms contain extracellular polymeric substances (EPS) that provide structural support and restrict penetration of antimicrobial treatment. To overcome limited penetration, functionalized nanoparticles (NPs) have been suggested as carriers for antimicrobial delivery. Using microscopy, we evaluate the diffusion of nanoparticles in function of the structure of Salmonella biofilms. We observe anomalous diffusion and heterogeneous mobility of NPs resulting in distinct NPs distribution that depended on biofilm structure. Through Brownian dynamics modeling with spatially varying viscosity around bacteria, we demonstrated that spatial gradients in diffusivity generate viscous sinks that trap NPs near bacteria. This model replicates the characteristic diffusion signature and vertical distribution of NPs in the biofilm. From a treatment perspective, our work indicates that both biofilm structure and the level of EPS can impact NP drug delivery, where low levels of EPS might benefit delivery by immobilizing NPs closer to bacteria and higher levels hamper delivery due to shielding effects.
Collapse
Affiliation(s)
- Bart Coppens
- Division of Mechatronics, Biostatistics, and Sensors, KU Leuven, 3001 Leuven, Belgium
| | - Tom E.R. Belpaire
- Division of Mechatronics, Biostatistics, and Sensors, KU Leuven, 3001 Leuven, Belgium
| | - Jiří Pešek
- Team SIMBIOTX, Inria Saclay, 91120 Palaiseau, France
| | | | - Herman Ramon
- Division of Mechatronics, Biostatistics, and Sensors, KU Leuven, 3001 Leuven, Belgium
| | - Bart Smeets
- Division of Mechatronics, Biostatistics, and Sensors, KU Leuven, 3001 Leuven, Belgium
| |
Collapse
|
43
|
Chung J, Eisha S, Park S, Morris AJ, Martin I. How Three Self-Secreted Biofilm Exopolysaccharides of Pseudomonas aeruginosa, Psl, Pel, and Alginate, Can Each Be Exploited for Antibiotic Adjuvant Effects in Cystic Fibrosis Lung Infection. Int J Mol Sci 2023; 24:ijms24108709. [PMID: 37240055 DOI: 10.3390/ijms24108709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/29/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
In cystic fibrosis (CF), pulmonary infection with Pseudomonas aeruginosa is a cause of increased morbidity and mortality, especially in patients for whom infection becomes chronic and there is reliance on long-term suppressive therapies. Current antimicrobials, though varied mechanistically and by mode of delivery, are inadequate not only due to their failure to eradicate infection but also because they do not halt the progression of lung function decline over time. One of the reasons for this failure is thought to be the biofilm mode of growth of P. aeruginosa, wherein self-secreted exopolysaccharides (EPSs) provide physical protection against antibiotics and an array of niches with resulting metabolic and phenotypic heterogeneity. The three biofilm-associated EPSs secreted by P. aeruginosa (alginate, Psl, and Pel) are each under investigation and are being exploited in ways that potentiate antibiotics. In this review, we describe the development and structure of P. aeruginosa biofilms before examining each EPS as a potential therapeutic target for combating pulmonary infection with P. aeruginosa in CF, with a particular focus on the current evidence for these emerging therapies and barriers to bringing these therapies into clinic.
Collapse
Affiliation(s)
- Jonathan Chung
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Shafinaz Eisha
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Subin Park
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Amanda J Morris
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
| | - Isaac Martin
- Department of Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, 686 Bay Street, Toronto, ON M5G 0A4, Canada
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
44
|
Saidi N, Davarzani F, Yousefpour Z, Owlia P. Effects of Sub-Minimum Inhibitory Concentrations of Gentamicin on Alginate Produced by Clinical Isolates of Pseudomonas aeruginosa. Adv Biomed Res 2023; 12:94. [PMID: 37288011 PMCID: PMC10241631 DOI: 10.4103/abr.abr_389_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 09/09/2022] [Accepted: 10/22/2022] [Indexed: 06/09/2023] Open
Abstract
Background Bacterial virulence factors may be influenced by sub-minimum inhibitory concentrations (sub-MICs) of antibiotics. The main purpose of this study was to investigate the effects of gentamicin at sub-MICs (0.5 MIC and 0.25 MIC) on alginate production of clinical isolates of Pseudomonas aeruginosa. Materials and Methods The minimum inhibitory concentrations of gentamicin against 88 clinical isolates of P. aeruginosa were determined using the broth microdilution method. Alginate production of the isolates in the absence and presence of gentamicin at sub-MICs was assessed by the carbazole method. The presence of alginate in clinical isolates was confirmed by the detection of alginate genes (algD and algU) using the PCR method. Results All the isolates had the ability of alginate production and were positive for algD and algU genes. sub-MICs of gentamicin significantly increased alginate production of 34 isolates (38.6%). On the other hand, in 49 isolates (55.7%), alginate production was significantly increased after treatment with sub-MICs of gentamicin. In five isolates (5.7%), the alginate production was reduced in exposure to 0.5 MIC of gentamicin while it was increased by gentamicin at 0.25 MIC. Conclusion This study showed different effects of gentamicin at sub-MICs on the alginate production of clinical isolates of P. aeruginosa. Further research is highly recommended to understand the mechanism of different responses of P. aeruginosa isolates to the exposure of sub-MICs of gentamicin.
Collapse
Affiliation(s)
- Navid Saidi
- Molecular Microbiology Research Center (MMRC), Faculty of Medicine, Shahed University, Tehran, Iran
| | - Fateme Davarzani
- Molecular Microbiology Research Center (MMRC), Faculty of Medicine, Shahed University, Tehran, Iran
| | - Zahra Yousefpour
- Molecular Microbiology Research Center (MMRC), Faculty of Medicine, Shahed University, Tehran, Iran
| | - Parviz Owlia
- Molecular Microbiology Research Center (MMRC), Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
45
|
Elmassry MM, Colmer-Hamood JA, Kopel J, San Francisco MJ, Hamood AN. Anti- Pseudomonas aeruginosa Vaccines and Therapies: An Assessment of Clinical Trials. Microorganisms 2023; 11:916. [PMID: 37110338 PMCID: PMC10144840 DOI: 10.3390/microorganisms11040916] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Pseudomonas aeruginosa is a Gram-negative opportunistic pathogen that causes high morbidity and mortality in cystic fibrosis (CF) and immunocompromised patients, including patients with ventilator-associated pneumonia (VAP), severely burned patients, and patients with surgical wounds. Due to the intrinsic and extrinsic antibiotic resistance mechanisms, the ability to produce several cell-associated and extracellular virulence factors, and the capacity to adapt to several environmental conditions, eradicating P. aeruginosa within infected patients is difficult. Pseudomonas aeruginosa is one of the six multi-drug-resistant pathogens (ESKAPE) considered by the World Health Organization (WHO) as an entire group for which the development of novel antibiotics is urgently needed. In the United States (US) and within the last several years, P. aeruginosa caused 27% of deaths and approximately USD 767 million annually in health-care costs. Several P. aeruginosa therapies, including new antimicrobial agents, derivatives of existing antibiotics, novel antimicrobial agents such as bacteriophages and their chelators, potential vaccines targeting specific virulence factors, and immunotherapies have been developed. Within the last 2-3 decades, the efficacy of these different treatments was tested in clinical and preclinical trials. Despite these trials, no P. aeruginosa treatment is currently approved or available. In this review, we examined several of these clinicals, specifically those designed to combat P. aeruginosa infections in CF patients, patients with P. aeruginosa VAP, and P. aeruginosa-infected burn patients.
Collapse
Affiliation(s)
- Moamen M. Elmassry
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Jane A. Colmer-Hamood
- Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jonathan Kopel
- Department of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Michael J. San Francisco
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Honors College, Texas Tech University, Lubbock, TX 79409, USA
| | - Abdul N. Hamood
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
46
|
Biofilm Formation and Control of Foodborne Pathogenic Bacteria. Molecules 2023; 28:molecules28062432. [PMID: 36985403 PMCID: PMC10058477 DOI: 10.3390/molecules28062432] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/17/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Biofilms are microbial aggregation membranes that are formed when microorganisms attach to the surfaces of living or nonliving things. Importantly, biofilm properties provide microorganisms with protection against environmental pressures and enhance their resistance to antimicrobial agents, contributing to microbial persistence and toxicity. Thus, bacterial biofilm formation is part of the bacterial survival mechanism. However, if foodborne pathogens form biofilms, the risk of foodborne disease infections can be greatly exacerbated, which can cause major public health risks and lead to adverse economic consequences. Therefore, research on biofilms and their removal strategies are very important in the food industry. Food waste due to spoilage within the food industry remains a global challenge to environmental sustainability and the security of food supplies. This review describes bacterial biofilm formation, elaborates on the problem associated with biofilms in the food industry, enumerates several kinds of common foodborne pathogens in biofilms, summarizes the current strategies used to eliminate or control harmful bacterial biofilm formation, introduces the current and emerging control strategies, and emphasizes future development prospects with respect to bacterial biofilms.
Collapse
|
47
|
Mechanisms and technology of marine oligosaccharides to control postharvest disease of fruits. Food Chem 2023; 404:134664. [DOI: 10.1016/j.foodchem.2022.134664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/18/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
|
48
|
Lin PY, Chan SY, Stern A, Chen PH, Yang HC. Epidemiological profiles and pathogenicity of Vancomycin-resistant Enterococcus faecium clinical isolates in Taiwan. PeerJ 2023; 11:e14859. [PMID: 36855433 PMCID: PMC9968458 DOI: 10.7717/peerj.14859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 01/16/2023] [Indexed: 02/25/2023] Open
Abstract
The emerging Vancomycin-resistant Enterococcus faecium (VRE-fm) is an opportunistic pathogen causing nosocomial infections. The identification of VRE-fm is important for successful prevention and control in healthcare settings. VRE-fm clinical isolates obtained from regional hospitals in northern Taiwan were characterized for antimicrobial susceptibility, virulence genes and biofilm production. Most isolates exhibited multi-drug resistance and carried the virulence genes, esp and hyl. While all isolates produce biofilms, those isolates that carried esp exhibited greater biofilm production. Isolates with different virulence gene carriages were examined for pathogenicity by using a nematode model, Caenorhabditis elegans, for determining microbial-host interactions. The survival assay showed that C. elegans was susceptible to Linezolid-resistant VRE-fm isolates with hyl. Combining the molecular epidemiological profiles regarding pathogenesis in C. elegans can serve as a guide for physicians in limiting opportunistic infections caused by VRE-fm.
Collapse
Affiliation(s)
- Pei-Yun Lin
- Department of Laboratory, Taipei City Hospital, Yang-Ming Branch, Taipei, Taiwan
| | - Shang-Yih Chan
- Department of Internal Medicine, Taipei City Hospital, Yang-Ming Branch, Taipei, Taiwan,Department of Exercise and Health Sciences, University of Taipei, Taipei, Taiwan,Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Arnold Stern
- Grossman School of Medicine, New York University, New York, USA
| | - Po-Hsiang Chen
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| |
Collapse
|
49
|
Kim YM, Son H, Park SC, Lee JK, Jang MK, Lee JR. Anti-Biofilm Effects of Rationally Designed Peptides against Planktonic Cells and Pre-Formed Biofilm of Pseudomonas aeruginosa. Antibiotics (Basel) 2023; 12:antibiotics12020349. [PMID: 36830260 PMCID: PMC9952520 DOI: 10.3390/antibiotics12020349] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023] Open
Abstract
Biofilms are resistant to antibiotics and are a major source of persistent and recurring infections by clinically important pathogens. Drugs used for biofilm-associated infections are limited because biofilm-embedded or biofilm-matrix bacteria are difficult to kill or eradiate. Therefore, many researchers are developing new and effective antibiofilm agents. Among them, antimicrobial peptides have an attractive interest in the development of antibiofilm agents. The present study evaluated the effects of 10 synthetic peptides on growth inhibition, inhibition of biofilm formation, and biofilm elimination in drug-resistant Pseudomonas aeruginosa. The planktonic cell growth and biofilm formation were dose-dependently inhibited by most of the peptides. WIK-14 eliminated preformed biofilm masses by removing carbohydrates, extracellular nucleic acids, proteins, and lipids constituting extracellular polymeric substances. The results demonstrated that WIK-14 and WIKE-14 peptides might provide novel therapeutic drugs to overcome multidrug resistance in biofilm-associated infections.
Collapse
Affiliation(s)
- Young-Min Kim
- Department of Chemical Engineering, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Hyosuk Son
- Department of Chemical Engineering, Sunchon National University, Suncheon 57922, Republic of Korea
- Department of Exhibition and Education, National Marine Biodiversity Institute of Korea, Seocheon 33662, Republic of Korea
| | - Seong-Cheol Park
- Department of Chemical Engineering, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Jong-Kook Lee
- Department of Chemical Engineering, Sunchon National University, Suncheon 57922, Republic of Korea
| | - Mi-Kyeong Jang
- Department of Chemical Engineering, Sunchon National University, Suncheon 57922, Republic of Korea
- Correspondence: (M.-K.J.); (J.R.L.)
| | - Jung Ro Lee
- LMO Team, National Institute of Ecology (NIE), Seocheon 33657, Republic of Korea
- Correspondence: (M.-K.J.); (J.R.L.)
| |
Collapse
|
50
|
Rouillard KR, Markovetz MR, Kissner WJ, Boone WL, Plott LM, Hill DB. Altering the viscoelastic properties of mucus-grown Pseudomonas aeruginosa biofilms affects antibiotic susceptibility. Biofilm 2023; 5:100104. [PMID: 36711323 PMCID: PMC9880403 DOI: 10.1016/j.bioflm.2023.100104] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023] Open
Abstract
The viscoelastic properties of biofilms are correlated with their susceptibility to mechanical and chemical stress, and the airway environment in muco-obstructive pulmonary diseases (MOPD) facilitates robust biofilm formation. Hyperconcentrated, viscoelastic mucus promotes chronic inflammation and infection, resulting in increased mucin and DNA concentrations. The viscoelastic properties of biofilms are regulated by biopolymers, including polysaccharides and DNA, and influence responses to antibiotics and phagocytosis. We hypothesize that targeted modulation of biofilm rheology will compromise structural integrity and increase antibiotic susceptibility and mucociliary transport. We evaluate biofilm rheology on the macro, micro, and nano scale as a function of treatment with a reducing agent, a biopolymer, and/or tobramycin to define the relationship between the viscoelastic properties of biofilms and susceptibility. Disruption of the biofilm architecture is associated with altered macroscopic and microscopic moduli, rapid vector permeability, increased antibiotic susceptibility, and improved mucociliary transport, suggesting that biofilm modulating therapeutics will improve the treatment of chronic respiratory infections in MOPD.
Collapse
Affiliation(s)
- Kaitlyn R. Rouillard
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Matthew R. Markovetz
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - William J. Kissner
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - William L. Boone
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Lucas M. Plott
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - David B. Hill
- Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Joint Department of Biomedical Engineering, North Carolina State University and the University of North Carolina, Chapel Hill, NC, 27599, USA,Department of Physics and Astronomy, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA,Corresponding author. Marsico Lung Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|